US5496938A - Nucleic acid ligands to HIV-RT and HIV-1 rev - Google Patents

Nucleic acid ligands to HIV-RT and HIV-1 rev Download PDF

Info

Publication number
US5496938A
US5496938A US07/964,624 US96462492A US5496938A US 5496938 A US5496938 A US 5496938A US 96462492 A US96462492 A US 96462492A US 5496938 A US5496938 A US 5496938A
Authority
US
United States
Prior art keywords
nucleic acid
hiv
ligand
seq
nucleic acids
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US07/964,624
Inventor
Larry Gold
Craig Tuerk
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Nexstar Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US07/714,131 external-priority patent/US5475096A/en
Application filed by Nexstar Pharmaceuticals Inc filed Critical Nexstar Pharmaceuticals Inc
Priority to US07/964,624 priority Critical patent/US5496938A/en
Assigned to NEXAGEN, INC. reassignment NEXAGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST. Assignors: GOLD, LARRY, TUERK, CRAIG
Priority to DE69333961T priority patent/DE69333961T2/en
Priority to ES93924904T priority patent/ES2257735T3/en
Priority to JP6509298A priority patent/JPH08501943A/en
Priority to EP93924904A priority patent/EP0668931B1/en
Priority to AU54411/94A priority patent/AU689087B2/en
Priority to EP05023827A priority patent/EP1683871B1/en
Priority to CA002145761A priority patent/CA2145761C/en
Priority to PT93924904T priority patent/PT668931E/en
Priority to AT05023827T priority patent/ATE518965T1/en
Priority to DK93924904T priority patent/DK0668931T3/en
Priority to PCT/US1993/009296 priority patent/WO1994008050A1/en
Priority to AT93924904T priority patent/ATE315101T1/en
Priority to US08/199,507 priority patent/US5472841A/en
Priority to US08/233,012 priority patent/US5849479A/en
Priority to US08/238,863 priority patent/US5503978A/en
Priority to US08/303,362 priority patent/US5648214A/en
Priority to US08/317,403 priority patent/US5629155A/en
Priority to US08/361,795 priority patent/US5756287A/en
Priority to US08/399,412 priority patent/US5622828A/en
Assigned to NEXSTAR PHARMCEUTICALS, INC. reassignment NEXSTAR PHARMCEUTICALS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: NEXAGEN, INC.
Priority to US08/430,709 priority patent/US5660985A/en
Priority to US08/433,585 priority patent/US5763566A/en
Priority to US08/433,126 priority patent/US5688935A/en
Priority to US08/434,425 priority patent/US5789157A/en
Priority to US08/434,001 priority patent/US5712375A/en
Priority to US08/433,124 priority patent/US5750342A/en
Priority to US08/437,667 priority patent/US5864026A/en
Priority to US08/441,591 priority patent/US5637682A/en
Priority to US08/442,062 priority patent/US5595877A/en
Priority to US08/441,828 priority patent/US5734034A/en
Priority to US08/442,572 priority patent/US5587468A/en
Priority to US08/443,407 priority patent/US5786462A/en
Priority to US08/447,172 priority patent/US5726017A/en
Priority to US08/447,169 priority patent/US5811533A/en
Priority to US08/458,423 priority patent/US5731144A/en
Priority to US08/458,424 priority patent/US5731424A/en
Priority to US08/465,591 priority patent/US5837834A/en
Priority to US08/465,594 priority patent/US5846713A/en
Priority to US08/471,985 priority patent/US5686592A/en
Priority to US08/479,783 priority patent/US5668264A/en
Priority to US08/484,557 priority patent/US5693502A/en
Priority to US08/479,725 priority patent/US5674685A/en
Priority to US08/487,720 priority patent/US5874557A/en
Priority to US08/487,425 priority patent/US5789163A/en
Priority to US08/477,527 priority patent/US5972599A/en
Priority to US08/488,402 priority patent/US5837456A/en
Priority to US08/484,552 priority patent/US5849890A/en
Priority to US08/477,530 priority patent/US5635615A/en
Priority to US08/477,830 priority patent/US5654151A/en
Priority to US08/472,256 priority patent/US6001988A/en
Priority to US08/479,729 priority patent/US5853984A/en
Priority to US08/479,724 priority patent/US5780228A/en
Priority to US08/481,710 priority patent/US6028186A/en
Priority to US08/487,426 priority patent/US5763173A/en
Priority to US08/472,255 priority patent/US5766853A/en
Publication of US5496938A publication Critical patent/US5496938A/en
Application granted granted Critical
Priority to US08/748,697 priority patent/US5817785A/en
Priority to US08/870,930 priority patent/US6168778B1/en
Priority to US08/918,304 priority patent/US5958691A/en
Priority to US08/976,413 priority patent/US6127119A/en
Priority to US09/046,247 priority patent/US6124449A/en
Priority to US09/165,616 priority patent/US6344318B1/en
Priority to US09/364,902 priority patent/US6232071B1/en
Priority to US09/363,939 priority patent/US6346611B1/en
Priority to US09/407,234 priority patent/US6184364B1/en
Assigned to GILEAD SCIENCES, INC. reassignment GILEAD SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEXSTAR PHARMACEUTICALS, INC.
Priority to US09/791,301 priority patent/US6713616B2/en
Priority to US09/854,662 priority patent/US6596491B2/en
Priority to US09/860,474 priority patent/US6696252B2/en
Priority to US10/040,497 priority patent/US6716583B2/en
Priority to JP2003411496A priority patent/JP2004097232A/en
Priority to US10/812,642 priority patent/US20040258656A1/en
Priority to US10/818,954 priority patent/US7368236B2/en
Priority to US11/286,221 priority patent/US20060084797A1/en
Priority to JP2008134843A priority patent/JP2008206524A/en
Priority to JP2009177035A priority patent/JP2009254380A/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/547Chelates, e.g. Gd-DOTA or Zinc-amino acid chelates; Chelate-forming compounds, e.g. DOTA or ethylenediamine being covalently linked or complexed to the pharmacologically- or therapeutically-active agent
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/22Tachykinins, e.g. Eledoisins, Substance P; Related peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1048SELEX
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1058Directional evolution of libraries, e.g. evolution of libraries is achieved by mutagenesis and screening or selection of mixed population of organisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6811Selection methods for production or design of target specific oligonucleotides or binding molecules
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/535Production of labelled immunochemicals with enzyme label or co-enzymes, co-factors, enzyme inhibitors or enzyme substrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/76Human chorionic gonadotropin including luteinising hormone, follicle stimulating hormone, thyroid stimulating hormone or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/13Decoys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/702Specific hybridization probes for retroviruses
    • C12Q1/703Viruses associated with AIDS
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F02COMBUSTION ENGINES; HOT-GAS OR COMBUSTION-PRODUCT ENGINE PLANTS
    • F02BINTERNAL-COMBUSTION PISTON ENGINES; COMBUSTION ENGINES IN GENERAL
    • F02B75/00Other engines
    • F02B75/02Engines characterised by their cycles, e.g. six-stroke
    • F02B2075/022Engines characterised by their cycles, e.g. six-stroke having less than six strokes per cycle
    • F02B2075/027Engines characterised by their cycles, e.g. six-stroke having less than six strokes per cycle four
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • G01N2333/163Regulatory proteins, e.g. tat, nef, rev, vif, vpu, vpr, vpt, vpx
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/50Fibroblast growth factors [FGF]
    • G01N2333/503Fibroblast growth factors [FGF] basic FGF [bFGF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/575Hormones
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/575Hormones
    • G01N2333/62Insulins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/81Protease inhibitors
    • G01N2333/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • G01N2333/811Serine protease (E.C. 3.4.21) inhibitors
    • G01N2333/8121Serpins
    • G01N2333/8125Alpha-1-antitrypsin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/916Hydrolases (3) acting on ester bonds (3.1), e.g. phosphatases (3.1.3), phospholipases C or phospholipases D (3.1.4)
    • G01N2333/918Carboxylic ester hydrolases (3.1.1)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • G01N2333/96436Granzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96433Serine endopeptidases (3.4.21)
    • G01N2333/96441Serine endopeptidases (3.4.21) with definite EC number
    • G01N2333/96455Kallikrein (3.4.21.34; 3.4.21.35)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/95Proteinases, i.e. endopeptidases (3.4.21-3.4.99)
    • G01N2333/964Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue
    • G01N2333/96425Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals
    • G01N2333/96427Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general
    • G01N2333/9643Proteinases, i.e. endopeptidases (3.4.21-3.4.99) derived from animal tissue from mammals in general with EC number
    • G01N2333/96486Metalloendopeptidases (3.4.24)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/966Elastase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/972Plasminogen activators
    • G01N2333/9726Tissue plasminogen activator
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/974Thrombin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/976Trypsin; Chymotrypsin

Definitions

  • Nucleic acid ligands are double or single stranded DNA or RNA species that bind specifically to a desired target molecule.
  • the basis for identifying nucleic acid ligands is a method that is called SELEX, an acronym for Systematic Evolution of Ligands by Exponential enrichment.
  • the methods of the present invention include means for analyzing and applying the information learned from the SELEX method to create an improved nucleic acid ligand for the selected target. These methods include computer modeling, boundary determination methods and chemical modification methods.
  • nucleic acid residues of a nucleic acid ligand are critical in binding to the selected target; 2) which nucleic acid residues affect the structural conformation of the nucleic acid ligand; and 3) what is the three-dimensional structure of the nucleic acid ligand.
  • This information allows for the identification and production of improved nucleic acid ligands that have superior binding capacity to the target as well as enhanced structural stability.
  • This information may also be utilized to produce non-nucleic acid or hybrid-nucleic acid species that also function as ligands to the target.
  • the methods of the present invention further provide an analysis of the target species that can be used in the preparation of therapeutic and/or diagnostic methods.
  • proteins or small molecules are not known to specifically bind to nucleic acids.
  • the known protein exceptions are those regulatory proteins such as repressors, polymerases, activators and the like which function in a living cell to bring about the transfer of genetic information encoded in the nucleic acids into cellular structures and the replication of the genetic material.
  • small molecules such as GTP bind to some intron RNAs.
  • nucleic acids either RNA or DNA
  • Crick both originally and in expanded form
  • nucleic acids can serve as templates for the synthesis of other nucleic acids through replicative processes that "read" the information in a template nucleic acid and thus yield complementary nucleic acids.
  • All of the experimental paradigms for genetics and gene expression depend on these properties of nucleic acids: in essence, double-stranded nucleic acids are informationally redundant because of the chemical concept of base pairs and because replicative processes are able to use that base pairing in a relatively error-free manner.
  • nucleic acids are thought to have narrower chemical possibilities than proteins, but to have an informational role that allows genetic information to be passed from virus to virus, cell to cell, and organism to organism.
  • nucleic acid components the nucleotides, must possess only pairs of surfaces that allow informational redundancy within a Watson-Crick base pair. Nucleic acid components need not possess chemical differences and activities sufficient for either a wide range of binding or catalysis.
  • nucleic acids found in nature do participate in binding to certain target molecules and even a few instances of catalysis have been reported.
  • the range of activities of this kind is narrow compared to proteins and more specifically antibodies.
  • nucleic acids are known to bind to some protein targets with high affinity and specificity, the binding depends on the exact sequences of nucleotides that comprise the DNA or RNA ligand.
  • short double-stranded DNA sequences are known to bind to target proteins that repress or activate transcription in both prokaryotes and eukaryotes.
  • Other short double-stranded DNA sequences are known to bind to restriction endonucleases, protein targets that can be selected with high affinity and specificity.
  • double-stranded DNA has a well-known capacity to bind within the nooks and crannies of target proteins whose functions are directed to DNA binding.
  • Single-stranded DNA can also bind to some proteins with high affinity and specificity, although the number of examples is rather smaller. From the known examples of double-stranded DNA binding proteins, it has become possible to describe the binding interactions as involving various protein motifs projecting amino acid side chains into the major groove of B form double-stranded DNA, providing the sequence inspection that allows specificity.
  • Double-stranded RNA occasionally serves as a ligand for certain proteins, for example, the endonuclease RNase III from E. coli.
  • RNase III the endonuclease from E. coli.
  • target proteins that bind to single-stranded RNA ligands, although in these cases the single-stranded RNA often forms a complex three-dimensional shape that includes local regions of intramolecular double-strandedness.
  • the amino-acyl tRNA synthetases bind tightly to tRNA molecules with high specificity.
  • a short region within the genomes of RNA viruses binds tightly and with high specificity to the viral coat proteins.
  • a short sequence of RNA binds to the bacteriophage T4-encoded DNA polymerase, again with high affinity and specificity.
  • RNA and DNA ligands either double- or single-stranded, serving as binding partners for specific protein targets.
  • Most known DNA binding proteins bind specifically to double-stranded DNA, while most RNA binding proteins recognize single-stranded RNA.
  • This statistical bias in the literature no doubt reflects the present biosphere's statistical predisposition to use DNA as a double-stranded genome and RNA as a single-stranded entity in the roles RNA plays beyond serving as a genome. Chemically there is no strong reason to dismiss single-stranded DNA as a fully able partner for specific protein interactions.
  • RNA and DNA have also been found to bind to smaller target molecules.
  • Double-stranded DNA binds to various antibiotics, such as actinomycin D.
  • a specific single-stranded RNA binds to the antibiotic thiostreptone; specific RNA sequences and structures probably bind to certain other antibiotics, especially those whose functions is to inactivate ribosomes in a target organism.
  • a family of evolutionary related RNAs binds with specificity and decent affinity to nucleotides and nucleosides (Bass, B. and Cech, T. (1984) Nature 308:820-826) as well as to one of the twenty amino acids (Yarus, M. (1988) Science 240:1751-1758).
  • Catalytic RNAs are now known as well, although these molecules perform over a narrow range of chemical possibilities, which are thus far related largely to phosphodiester transfer reactions and hydrolysis of nucleic acids.
  • nucleic acids as chemical compounds can form a virtually limitless array of shapes, sizes and configurations, and are capable of a far broader repertoire of binding and catalytic functions than those displayed in biological systems.
  • RNA site of bacteriophage R17 coat protein binding has been identified by Uhlenbeck and coworkers.
  • the minimal natural RNA binding site (21 bases long) for the R17 coat protein was determined by subjecting variable-sized labeled fragments of the mRNA to nitrocellulose filter binding assays in which protein-RNA fragment complexes remain bound to the filter (Carey et al. (1983) Biochemistry 22:2601).
  • a number of sequence variants of the minimal R17 coat protein binding site were created in vitro in order to determine the contributions of individual nucleic acids to protein binding (Uhlenbeck et al. (1983) J. Biomol.
  • the Q ⁇ coat protein RNA binding site was found to be similar to that of R17 in size, and in predicted secondary structure, in that it comprised about 20 bases with an 8 base pair hairpin structure which included a bulged nucleotide and a 3 base loop. In contrast to the R17 coat protein binding site, only one of the single-stranded residues of the loop is essential for binding and the presence of the bulged nucleotide is not required.
  • the protein-RNA binding interactions involved in translational regulation display significant specificity.
  • Nucleic acids are known to form secondary and tertiary structures in solution.
  • the double-stranded forms of DNA include the so-called B double-helical form, Z-DNA and superhelical twists (Rich, A. et al. (1984) Ann. Rev. Biochem. 53:791-846).
  • Single-stranded RNA forms localized regions of secondary structure such as hairpin loops and pseudoknot structures (Schimmel, P. (1989) Cell 58:9-12).
  • the phage RNA serves as a poly-cistronic messenger RNA directing translation of phage-specific proteins and also as a template for its own replication catalyzed by Q ⁇ RNA replicase.
  • This RNA replicase was shown to be highly specific for its own RNA templates.
  • RNAs were isolated which were also replicated by Q ⁇ replicase. Minor alterations in the conditions under which cycles of replication were performed were found to result in the accumulation of different RNAs, presumably because their replication was favored under the altered conditions.
  • RNA had to be bound efficiently by the replicase to initiate replication and had to serve as a kinetically favored template during elongation of RNA.
  • Kramer et al. (1974) J. Mol. Biol. 89:719 reported the isolation of a mutant RNA template of Q ⁇ replicase, the replication of which was more resistant to inhibition by ethidium bromide than the natural template. It was suggested that this mutant was not present in the initial RNA population but was generated by sequential mutation during cycles of in vitro replication with Q ⁇ replicase. The only source of variation during selection was the intrinsic error rate during elongation by Q ⁇ replicase.
  • selection occurred by preferential amplification of one or more of a limited number of spontaneous variants of an initially homogenous RNA sequence. There was no selection of a desired result, only that which was intrinsic to the mode of action of Q ⁇ replicase.
  • RNAs which specifically cleave single-stranded DNA reported a method for identifying RNAs which specifically cleave single-stranded DNA.
  • the selection for catalytic activity was based on the ability of the ribozyme to catalyze the cleavage of a substrate ssRNA or DNA at a specific position and transfer the 3'-end of the substrate to the 3'-end of the ribozyme.
  • the product of the desired reaction was selected by using an oligodeoxynucleotide primer which could bind only to the completed product across the junction formed by the catalytic reaction and allowed selective reverse transcription of the ribozyme sequence.
  • the selected catalytic sequences were amplified by attachment of the promoter of T7 RNA polymerase to the 3'-end of the cDNA, followed by transcription to RNA. The method was employed to identify from a small number of ribozyme variants the variant that was most reactive for cleavage of a selected substrate.
  • the method of the SELEX Patent Applications is based on the unique insight that nucleic acids have sufficient capacity for forming a variety of two- and three-dimensional structures and sufficient chemical versatility available within their monomers to act as ligands (form specific binding pairs) with virtually any chemical compound, whether large or small in size.
  • the method involves selection from a mixture of candidates and step-wise iterations of structural improvement, using the same general selection theme, to achieve virtually any desired criterion of binding affinity and selectivity.
  • SELEX a mixture of nucleic acids, preferably comprising a segment of randomized sequence
  • the method includes steps of contacting the mixture with the target under conditions favorable for binding, partitioning unbound nucleic acids from those nucleic acids which have bound to target molecules, dissociating the nucleic acid-target pairs, amplifying the nucleic acids dissociated from the nucleic acid-target pairs to yield a ligand-enriched mixture of nucleic acids, then reiterating the steps of binding, partitioning, dissociating and amplifying through as many cycles as desired.
  • SELEX is based on the inventors' insight that within a nucleic acid mixture containing a large number of possible sequences and structures there is a wide range of binding affinities for a given target.
  • a nucleic acid mixture comprising, for example a 20 nucleotide randomized segment can have 4 20 candidate possibilities. Those which have the higher affinity constants for the target are most likely to bind.
  • a second nucleic acid mixture is generated, enriched for the higher binding affinity candidates. Additional rounds of selection progressively favor the best ligands until the resulting nucleic acid mixture is predominantly composed of only one or a few sequences. These can then be cloned, sequenced and individually tested for binding affinity as pure ligands.
  • the method may be used to sample as many as about 10 18 different nucleic acid species.
  • the nucleic acids of the test mixture preferably include a randomized sequence portion as well as conserved sequences necessary for efficient amplification.
  • Nucleic acid sequence variants can be produced in a number of ways including synthesis of randomized nucleic acid sequences and size selection from randomly cleaved cellular nucleic acids.
  • the variable sequence portion may contain fully or partially random sequence; it may also contain subportions of conserved sequence incorporated with randomized sequence. Sequence variation in test nucleic acids can be introduced or increased by mutagenesis before or during the selection/amplification iterations.
  • the selection process is so efficient at isolating those nucleic acid ligands that bind most strongly to the selected target, that only one cycle of selection and amplification is required.
  • Such an efficient selection may occur, for example, in a chromatographic-type process wherein the ability of nucleic acids to associate with targets bound on a column operates in such a manner that the column is sufficiently able to allow separation and isolation of the highest affinity nucleic acid ligands.
  • the target-specific nucleic acid ligand solution may include a family of nucleic acid structures or motifs that have a number of conserved sequences and a number of sequences which can be substituted or added without significantly effecting the affinity of the nucleic acid ligands to the target.
  • nucleic acid primary, secondary and tertiary structures are known to exist.
  • the structures or motifs that have been shown most commonly to be involved in non-Watson-Crick type interactions are referred to as hairpin loops, symmetric and asymmetric bulges, psuedoknots and myriad combinations of the same.
  • Almost all known cases of such motifs suggest that they can be formed in a nucleic acid sequence of no more than 30 nucleotides. For this reason, it is often preferred that SELEX procedures with contiguous randomized segments be initiated with nucleic acid sequences containing a randomized segment of between about 20-50 nucleotides.
  • the SELEX Patent Applications also describe methods for obtaining nucleic acid ligands that bind to more than one site on the target molecule, and to nucleic acid ligands that include non-nucleic acid species that bind to specific sites on the target.
  • the SELEX method provides means for isolating and identifying nucleic acid ligands which bind to any envisonable target.
  • the SELEX method is applied to situations where the target is a protein, including both nucleic acid-binding proteins and proteins not known to bind nucleic acids as part of their biological function.
  • RNA structure at high resolution Little is known about RNA structure at high resolution.
  • the basic A-form helical structure of double stranded RNA is known from fiber diffraction studies.
  • X-ray crystallography has yielded the structure of a few tRNAs and a short poly-AU helix.
  • the X-ray structure of a tRNA/synthetase RNA/protein complex has also been solved.
  • the structures of two tetranucleotide hairpin loops and one model pseudoknot are know from NMR studies.
  • RNAs often appear to make well-defined secondary structures which provide more stability than the tertiary interactions.
  • the tertiary structure of tRNA can be disrupted without disrupting the secondary structure by chelation of magnesium or by raising the temperature.
  • Secondary structure prediction for RNAs is well-understood, and is generally quite accurate for small RNA molecules. For RNAs, structural prediction can be broken into subproblems; first, predict the secondary structure; then, predict how the resulting helices and remaining single strands are arranged relative to each other.
  • RNA For RNA, the first attempts at structural prediction were for tRNAs.
  • the secondary structure of the canonical tRNA cloverleaf was known from comparative sequence analysis, reducing the problem to one of arranging four short A-form helices in space relative to each other.
  • Manual CPK modeling, back-of-the-envelope energy minimization, and a few distance restraints available from crosslinking studies and phylogenetic covariations were used to generate a tRNA model--which unfortunately proved wrong when the first crystal structure of phenylalanine tRNA was solved a few years later.
  • Computer modeling has supplanted manual modeling, relieving the model-builder of the difficulties imposed by gravitation and mass.
  • Computer modeling can only be used without additional experimental data for instances in which a homologous structure is known; for instance, the structure of the 3' end of the turnip yellow mosaic virus RNA genome was modeled, based on the known 3D structure of tRNA and the knowledge that the 3' end of TYMV is recognized as tRNA-like by a number of cellular tRNA modification enzymes.
  • This model was the first 3D model of an RNA pseudoknot; the basic structure of an isolated model pseudoknot has been corroborated by NMR data.
  • Francois Michel ((1989) Nature 342:391) has constructed a model for the catalytic core of group I introns. Like the tRNAs, the secondary structure of group I intron cores is well-known from comparative sequence analysis, so the problem is reduced to one of properly arranging helices and the remaining single-stranded regions. Michel ((1989) supra) analyzed an aligned set of 87 group I intron sequences by eye and detected seven strong pairwise and triplet covariations outside of the secondary structure, which he interpreted as tertiary contacts and manually incorporated as restraints on his model. As yet, there is no independent confirmation of the Michel model.
  • RNA is treated as an assemblage of cylinders (A-form helices) and beads (single-stranded residues), and a mathematical technique called distance geometry is used to generate arrangements of these elements which are consistent with a set of distance restraints.
  • this protocol Using a small set of seven distance restraints on the phenylalanine tRNA tertiary structure, this protocol generated the familiar L-form of the tRNA structure about 2/3 of the time.
  • the present invention includes methods for identifying and producing nucleic acid ligands and the nucleic acid ligands so identified and produced.
  • the SELEX method described above allows for the identification of a single nucleic acid ligand or a family of nucleic acid ligands to a given target.
  • the methods of the present invention allow for the analysis of the nucleic acid ligand or family of nucleic acid ligands obtained by SELEX in order to identify and produce improved nucleic acid ligands.
  • nucleic acid ligands include mathematical modeling and structure modifications of the SELEX derived ligands. Further included are methods for determining which nucleic acid residues in a nucleic acid ligand are necessary for maintaining the three-dimensional structure of the ligand, and which residues interact with the target to facilitate the formation of ligand-target binding pairs.
  • nucleic acid ligands are desired for their ability to inhibit one or more of the biological activities of the target. In such cases, methods are provided for determining whether the nucleic acid ligand effectively inhibits the desired biological activity.
  • the present invention includes improved nucleic acid ligands to the HIV-RT and HIV-1 Rev proteins. Also included are nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind HIV-RT or the HIV-1 Rev protein as the nucleic acid ligands specifically identified herein.
  • Also included within the scope of the invention is a method for performing sequential SELEX experiments in order to identify extended nucleic acid ligands.
  • extended nucleic acid ligands to the HIV-RT protein are disclosed.
  • Nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind HIV-RT as the extended HIV-RT nucleic acid ligands are also included in this invention.
  • FIG. 1 depicts the consensus pseudoknot derived from primary and secondary SELEX experiments describing high affinity inhibitory ligands of HIV-1 reverse transcriptase (HIV-RT).
  • the consensus secondary structure is a pseudoknot; the 5' helix of that pseudoknot (Stem 1) is conserved at the primary sequence level and the 3' helix or Stem 2 is not.
  • X indicates a nucleotide position that is non-conserved; X-X' indicates a preferred base-pair. The 26 nucleotide positions are numbered as shown.
  • FIG. 2A depicts refinement of the 5' information boundary.
  • a set of model ligands was synthesized with T7 RNA polymerase from template oligos. Milligan et al. Nucl. Acid. Res., 15:8783-8798 (1987). Illustrated in the upper left is the complete ligand B. On the right margin are shown the variations in the individual ligands A through E that occur in the boxed areas.
  • FIG. 2B depicts graphically the :individual binding curves for these model ligands.
  • FIG. 3 depicts the effect of various nucleotide substitutions within the ligand B sequence on binding to HIV-RT. Illustrated are the various substitutions and resultant affinities to HIV-RT expressed relative to the binding of ligand B.
  • Ligand B was a control tested in each experiment; the affinity of ligand B is normalized as 1.0 and the relative affinity (Kd of ligand B is divided by the Kd of each ligand) is shown. Also shown are the affinities of various truncations of ligand B. The value associated with the asterisked G--G which replaces U1-G16 comes from ligand C of FIG. 2.
  • FIG. 4 depicts a chemical probe of the native versus denatured conformations of ligand B.
  • the various nucleotides of ligand B were reacted with chemicals under native and denaturing conditions, assayed for the modified positions, electrophoresed and visualized for comparison. indicate highly reactive base-pairing groups of the base at that position and ⁇ partially reactivity; indicates strong reactivity of purine N7 positions and .increment. partial reactivity (to modification with DEPC).
  • the question marks indicate that these positions on G(-2) and G(-1) could not be distinguished due to band crowding on the gel.
  • FIG. 5 depicts reactivities of modifiable groups of ligand B when bound to HIV-RT. Diagrammed are those groups that show altered reactivity when bound to HIV-RT as compared to that of the native conformation.
  • FIG. 6 depicts modification interference results for ligand B complexing with HIV-RT. Symbols for modification are as in the boxed legend. The modifications indicated are those that are strongly (filled symbols) or partially (unfilled symbols) selected against by binding to HIV-RT (reflected by decreased modification at those positions in the selected population).
  • FIG. 7A depicts substitution of 2'-methoxy for hydroxyl on the riboses of the ligand B sequence shown in the upper right. Illustrated in the upper right is the complete ligand B. On the right margin are shown the variations in the individual ligands A through D that occur in ligand B.
  • FIG. 7B depicts graphically the individual binding curves for ligands A-D.
  • FIG. 8 depicts selection by HIV-RT from mixed populations of 2'-methoxy ribose versus 2'-hydroxyl at positions U1 through A5 and A12 through A20.
  • An oligonucleotide was synthesized with the following sequence:
  • subscripted "d” indicates 2'-deoxy
  • subscripted "x” that those nucleotides are mixed 50--50 for phosphoramidite reagents resulting in 2'-methoxy or 2'-hydroxyl on the ribose
  • subscripted "m” indicating that those nucleotides are all 2'-methoxy on the ribose.
  • FIG. 9 shows the starting RNA (SEQ ID NO:37) and the collection of sequences, grouped into two motifs, Extension Motif I (SEQ ID NOS: 14-27) and Extension Motif II (SEQ ID NO8:28-33), obtained from SELEX with HIV-RT as part of a walking experiment.
  • FIG. 10 illustrates the consensus extended HIV-RT ligand obtained from the list of sequences shown in FIG. 9.
  • FIG. 10A illustrates the secondary structure of the first 25 bases of the starting material (SEQ ID NO:37) shown in FIG. 9.
  • FIGS. 10B and 10C illustrate the consensus extended HIV-RT ligands obtained from the list of sequences, Extension Motif I (SEQ ID NOS:14-27) (FIG. 10B) and Extension Motif II (SEQ ID NO8:28-33) (FIG. 10C), shown in FIG. 9.
  • FIG. 11 illustrates the revised description of the pseudoknot ligand of HIV-RT.
  • the S-S' indicates the preferred C-G or G-C base-pair at this position.
  • FIG. 12A shows the sequence of a high-affinity RNA ligand for HIV-1 Rev protein obtained from SELEX experiments. Shown is the numbering scheme used for reference to particular bases in the RNA. This sequence was used for chemical modification with ENU.
  • FIG. 12B shows the extended RNA sequence used in chemical modification experiments with DMS, kethoxal, CMCT, and DEPC.
  • FIG. 12C shows the sequence of the oligonucleotide used for primer extension of the extended ligand sequence.
  • FIG. 13 depicts the results of chemical modification of the HIV-1 Rev ligand RNA under native conditions
  • a) lists chemical modifying agents, their specificity, and the symbols denoting partial and full modification.
  • the RNA sequence is shown, with degree and type of modification displayed for every modified base
  • b) depicts the helical, bulge, and hairpin structural elements of the HIV-1 Rev RNA ligand corresponding to the modification and computer structural prediction data.
  • FIG. 14 depicts the results of chemical modification of the ligand RNA that interferes with binding to the HIV-1 Rev protein. Listed are the modifications which interfere with protein binding, classified into categories of strong interference and slight interference. Symbols denote either base-pairing modifications, N7 modifications, or phosphate modifications.
  • FIG. 15 depicts the modification interference values for phosphate alkylation. Data is normalized to A17 3' phosphate.
  • FIG. 16 depicts the modification interference values for DMS modification of N3C and N1A. Data is normalized to C36; A34.
  • FIG. 17 depicts the modification interference values for kethoxal modification of N1G and N2G. Data is normalized to G5.
  • FIG. 18 depicts the modification interference values for CMCT modification of N3U and N1G. Data is normalized to U38.
  • FIG. 19 depicts the modification interference values for DEPC modification of N7A and N7G. Data normalized to G19; A34.
  • FIG. 20 depicts the chemical modification of the RNA ligand in the presence of the HIV-1 Rev protein. Indicated are those positions that showed either reduced modification or enhanced modification in the presence of protein as compared to modification under native conditions but without protein present.
  • FIG. 21 shows the 5' and 3' sequences which flank the "6a" biased random region used in SELEX.
  • the template which produced the initial RNA population was constructed from the following oligonucleotides:
  • the small-case letters in the template oligo indicate that at each position that a mixture of reagents were used in synthesis by an amount of 62.5% of the small case letter, and 12.5% each of the other three nucleotides.
  • Listed below the 6a sequence are the sequences of 38 isolates cloned after six rounds of SELEX performed with Rev protein with this population of RNA. The differences found in these isolates from the 6a sequences are indicated by bold-faced characters. Underlined are the predicted base pairings that comprise the bulge-flanking stems of the Motif I Rev ligands. Bases that are included from the 5' and 3' fixed flanking sequences are lower case.
  • FIG. 22 shows three sets of tabulations containing:
  • FIG. 23 shows three sets of tabulations containing:
  • C) The difference between the fractional frequency of B) and the expected frequency based on the input mixture of oligonucleotides during template synthesis [for "wild type" positions, (x ⁇ 38)-0.39; for base pairs that contain one alternate nucleotide and one wild type nucleotide, (x ⁇ 38)-0.078; and for base pairings of two alternate nucleotides (x ⁇ 38)-0.016]. Values are shown for purine pyrimidine pairings only, the other eight pyrimidine and purine pairings are collectively counted and shown as "other" and are computed for section
  • FIG. 24A shows the previously determined Rev protein ligand Motif I consensus from U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991.
  • FIG. 24B shows the 6a sequence from the same application.
  • FIG. 24C shows the preferred consensus derived from the biased randomization SELEX as interpreted from the data presented in FIGS. 22 and 23.
  • S-S' indicates either a C-G or G-C base pair.
  • This application includes methods for identifying and producing improved nucleic acid ligands based on the basic SELEX process.
  • the application includes separate sections covering the following embodiments of the invention: I. The SELEX Process; II. Techniques for Identifying Improved Nucleic Acid Ligands Subsequent to Performing SELEX; III. Sequential SELEX Experiments--Walking; IV. Elucidation of Structure of Ligands Via Covariance Analysis; V. Elucidation of an Improved Nucleic Acid Ligand for HIV-RT; VI. Performance of Walking Experiment With HIV-RT Nucleic Acid Ligand to Identify Extended Nucleic Acid Ligands; and VII. Elucidation of an Improved Nucleic Acid Ligand for HIV-1 Rev Protein.
  • nucleic acid ligands to the HIV-RT and HIV-1 Rev proteins are disclosed and claimed herein.
  • This invention includes the specific nucleic acid ligands identified herein. The scope of the ligands covered by the invention extends to all ligands of the HIV-RT and Rev proteins identified according to the procedures described herein. More specifically, this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind the HIV-RT or Rev proteins, under physiological conditions, as the nucleic acid ligands identified herein. By substantially homologous, it is meant, a degree of homology in excess of 70%, most preferably in excess of 80%.
  • Substantially homologous also includes base pair flips in those areas of the nucleic acid ligands that include base pairing regions.
  • Substantially the same ability to bind the HIV-RT or Rev protein means that the affinity is within two orders of magnitude of the affinity of the nucleic acid ligands described herein. It is well within the skill of those of ordinary skill in the art to determine whether a given sequence is substantially homologous to and has substantially the same ability to bind the HIV-RT or HIV-1 Rev protein as the sequences identified herein.
  • the SELEX process may be defined by the following series of steps:
  • a candidate mixture of nucleic acids of differing sequence is prepared.
  • the candidate mixture generally includes regions of fixed sequences (i.e., each of the members of the candidate mixture contains the same sequences in the same location) and regions of randomized sequences.
  • the fixed sequence regions are selected either: a) to assist in the amplification steps described below; b) to facilitate mimicry of a sequence known to bind to the target; or c) to enhance the concentration of a given structural arrangement of the nucleic acids in the candidate mixture.
  • the randomized sequences can be totally randomized (i.e., the probability of finding a base at any position being one in four) or only partially randomized (e.g., the probability of finding a base at any location can be selected at any level between 0 and 100 percent).
  • the candidate mixture is contacted with the selected target under conditions favorable for binding between the target and members of the candidate mixture. Under these circumstances, the interaction between the target and the nucleic acids of the candidate mixture can be considered as forming nucleic acid-target pairs between the target and the nucleic acids having the strongest affinity for the target.
  • nucleic acids with the highest affinity for the target are partitioned from those nucleic acids with lesser affinity to the target. Because only an extremely small number of sequences (and possibly only one molecule of nucleic acid) corresponding to the highest affinity nucleic acids exist in the candidate mixture, it is generally desirable to set the partitioning criteria so that a significant amount of the nucleic acids in the candidate mixture (approximately 5-50%) are retained during partitioning.
  • nucleic acids selected during partitioning as having the relatively higher affinity to the target are then amplified to create a new candidate mixture that is enriched in nucleic acids having a relatively higher affinity for the target.
  • the newly formed candidate mixture contains fewer and fewer unique sequences, and the average degree of affinity of the nucleic acids to the target will generally increase.
  • the SELEX process will yield a candidate mixture containing one or a small number of unique nucleic acids representing those nucleic acids from the original candidate mixture having the highest affinity to the target molecule.
  • the SELEX Patent Applications describe and elaborate on this process in great detail. Included are targets that can be used in the process; methods for the preparation of the initial candidate mixture; methods for partitioning nucleic acids within a candidate mixture; and methods for amplifying partitioned nucleic acids to generate enriched candidate mixtures.
  • the SELEX Patent Applications also describe ligand solutions obtained to a number of target species, including both protein targets wherein the protein is and is not a nucleic acid binding protein.
  • SELEX delivers high affinity ligands of a target molecule. This represents a singular achievement that is unprecedented in the field of nucleic acids research.
  • the present invention is directed at methods for taking the SELEX derived ligand solution in order to develop novel nucleic acid ligands having the desired characteristics.
  • the desired characteristics for a given nucleic acid ligand may vary. All nucleic acid ligands are capable of forming a complex with the target species. In some cases, it is desired that the nucleic acid ligand will serve to inhibit one or more of the biological activities of the target. In other cases, it is desired that the nucleic acid ligand serves to modify one or more of the biological activities of the target. In other cases, the nucleic acid ligand serves to identify the presence of the target, and its effect on the biological activity of the target is irrelevant.
  • the nucleic acid ligand 1) binds to the target in a manner capable of achieving the desired effect on the target; 2) be as small as possible to obtain the desired effect; 3) be as stable as possible; and 4) be a specific ligand to the chosen target. In most, if not all, situations it is preferred that the nucleic acid ligand have the highest possible affinity to the target. Modifications or derivatizations of the ligand that confer resistance to degradation and clearance in situ during therapy, the capability to cross various tissue or cell membrane barriers, or any other accessory properties that do not significantly interfere with affinity for the target molecule may also be provided as improvements.
  • the present invention includes the methods for obtaining improved nucleic acid ligands after SELEX has been performed.
  • Assays of ligand effects on target molecule function One of the uses of nucleic acid ligands derived by SELEX is to find ligands that alter target molecule function. Because ligand analysis requires much more work than is encountered during SELEX enrichments, it is a good procedure to first assay for inhibition or enhancement of function of the target protein. One could even perform such functional tests of the combined ligand pool prior to cloning and sequencing. Assays for the biological function of the chosen target are generally available and known to those skilled in the art, and can be easily performed in the presence of the nucleic acid ligand to determine if inhibition occurs.
  • Affinity assays of the ligands SELEX enrichment will supply a number of cloned ligands of probable variable affinity for the target molecule. Sequence comparisons may yield consensus secondary structures and primary sequences that allow grouping of the ligand sequences into motifs. Although a single ligand sequence (with some mutations) can be found frequently in the total population of cloned sequences, the degree of representation of a single ligand sequence in the cloned population of ligand sequences may not absolutely correlate with affinity for the target molecule.
  • Information boundaries determination An important avenue for narrowing down what amount of sequence is relevant to specific affinity is to establish the boundaries of that information within a ligand sequence. This is conveniently accomplished by selecting end-labeled fragments from hydrolyzed pools of the ligand of interest so that 5' and 3' boundaries of the information can be discovered. To determine a 3' boundary, one performs a large-scale in vitro transcription of the PCRd ligand, gel purifies the RNA using UV shadowing on an intensifying screen, phosphatases the purified RNA, phenol extracts extensively, labels by kinasing with 32P, and gel purifies the labeled product (using a film of the gel as a guide).
  • the resultant product may then be subjected to pilot partial digestions with RNase T1 (varying enzyme concentration and time, at 50° C. in a buffer of 7M urea, 50 mM NaCitrate pH 5.2) and alkaline hydrolysis (at 50 mM NaCO 3 , adjusted to pH 9.0 by prior mixing of 1M bicarbonate and carbonate solutions; test over ranges of 20 to 60 minutes at 95° C.).
  • RNase T1 varying enzyme concentration and time, at 50° C. in a buffer of 7M urea, 50 mM NaCitrate pH 5.2
  • alkaline hydrolysis at 50 mM NaCO 3 , adjusted to pH 9.0 by prior mixing of 1M bicarbonate and carbonate solutions; test over ranges of 20 to 60 minutes at 95° C.
  • target concentration if target supplies allow
  • the RNA is eluted as in SELEX and then run on a denaturing gel with T1 partial digests so that the positions of hydrolysis bands can be related to the ligand sequence.
  • the 5' boundary can be similarly determined.
  • Large-scale in vitro transcriptions are purified as described above. There are two methods for labeling the 3' end of the RNA.
  • One method is to kinase Cp with 32P (or purchase 32P-Cp) and ligate to the purified RNA with RNA ligase.
  • the labeled RNA is then purified as above and subjected to very identical protocols.
  • An alternative is to subject unlabeled RNAs to partial alkaline hydrolyses and extend an annealed, labeled primer with reverse transcriptase as the assay for band positions.
  • RNA extension on eluted RNA sometimes contains artifactual stops, so it may be important to control by spotting and eluting starting material on nitrocellulose filters without washes and assaying as the input RNA.
  • the result is that it is possible to find the boundaries of the sequence information required for high affinity binding to the target.
  • the highest affinity ligands contained the essential information UCCGNNNNNNNNCGGGAAAAN'N'N'N' (SEQ ID NO:2) (where N's base pair to Ns in the 8 base loop sequence of the hairpin formed by the pairing of UCCG to CGGG) and that the 5' U would be dispensable with some small loss in affinity.
  • the construction of model compounds confirmed that there was no difference in the affinity of sequences with only one 5' U compared to 2 5' U's (as is shared by the two compared ligands), that removal of both U's caused a 5-fold decrease in affinity and of the next C a more drastic loss in affinity.
  • SECONDARY SELEX Once the minimal high affinity ligand sequence is identified, it may be useful to identify the nucleotides within the boundaries that are crucial to the interaction with the target molecule.
  • One method is to create a new random template in which all of the nucleotides of a high affinity ligand sequence are partially randomized or blocks of randomness are interspersed with blocks of complete randomness.
  • Such "secondary" SELEXes produce a pool of ligand sequences in which crucial nucleotides or structures are absolutely conserved, less crucial features preferred, and unimportant positions unbiased. Secondary SELEXes can thus help to further elaborate a consensus that is based on relatively few ligand sequences. In addition, even higher-affinity ligands may be provided whose sequences were unexplored in the original SELEX.
  • this sequence was incorporated in a secondary SELEX experiment in which each of the nucleotides of the 6a sequence (confined to that part of the sequence which comprises a Rev protein binding site defined by homology to others of Rev ligand motif I) was mixed during oligonucleotide synthesis with the other three nucleotides in the ratio 62.5:12.5:12.5:12.5.
  • the reagents for G,A,T, and C are mixed in the ratios 62.5:12.5:12.5:12.5.
  • ligands were cloned from this mixture so that a more comprehensive consensus description could be derived.
  • NUCLEOTIDE SUBSTITUTION Another method is to test oligo-transcribed variants where the SELEX consensus may be confusing. As shown above, this has helped us to understand the nature of the 5' and 3' boundaries of the information required to bind HIV-RT. As is shown in the attached example this has helped to quantitate the consensus of nucleotides within Stem 1 of the HIV-RT pseudoknot.
  • CHEMICAL MODIFICATION Another useful set of techniques are inclusively described as chemical modification experiments. Such experiments may be used to probe the native structure of RNAs, by comparing modification patterns of denatured and non-denatured states. The chemical modification pattern of an RNA ligand that is subsequently bound by target molecule may be different from the native pattern, indicating potential changes in structure upon binding or protection of groups by the target molecule. In addition, RNA ligands will fail to be bound by the target molecule when modified at positions crucial to either the bound structure of the ligand or crucial to interaction with the target molecule. Such experiments in which these positions are identified are described as "chemical modification interference" experiments.
  • Ethylnitrosourea modification interference identified 5 positions at which modification interfered with binding and 2 of those positions at which it interfered drastically. Modification of various atomic groups on the bases of the ligand were also identified as crucial to the interaction with HIV-RT. Those positions were primarily in the 5' helix and bridging loop sequence that was highly conserved in the SELEX phylogeny (Stem I and Loop 2, FIG. 1). These experiments not only confirmed the validity of that phylogeny, but informed ongoing attempts to make more stable RNAs.
  • RT ligand was synthesized in which all positions had 2'-methoxy at the ribose portions of the backbone. This molecule bound with drastically reduced affinity for HIV-RT. Based on the early modification interference experiments and the SELEX phylogeny comparisons, it could be determined that the 3' helix (Stem II FIG. 1) was essentially a structural component of the molecule. A ligand in which the 12 ribose residues of that helix were 2'-methoxy was then synthesized and it bound with high affinity to HIV-RT.
  • Comparisons of the intensity of bands for bound and unbound ligands may reveal not only modifications that interfere with binding, but also modifications that enhance binding.
  • a ligand may be made with precisely that modification and tested for the enhanced affinity.
  • chemical modification experiments can be a method for exploring additional local contacts with the target molecule, just as “walking” (see below) is for additional nucleotide level contacts with adjacent domains.
  • One of the products of the SELEX procedure is a consensus of primary and secondary structures that enables the chemical or enzymatic synthesis of oligonucleotide ligands whose design is based on that consensus. Because the replication machinery of SELEX requires that rather limited variation at the subunit level (ribonucleotides, for example), such ligands imperfectly fill the available atomic space of a target molecule's binding surface. However, these ligands can be thought of as high-affinity scaffolds that can be derivatized to make additional contacts with the target molecule. In addition, the consensus contains atomic group descriptors that are pertinent to binding and atomic group descriptors that are coincidental to the pertinent atomic group interactions.
  • each ribonucleotide of the pseudoknot ligand of HIV-RT contains a 2' hydroxyl group on the ribose, but only two of the riboses of the pseudoknot ligand cannot be substituted at this position with 2'-methoxy.
  • a similar experiment with deoxyribonucleotide mixtures with ribonucleotide mixtures would reveal which riboses or how many riboses are dispensable for binding HIV-RT.
  • a similar experiment with more radical substitutions at the 2' position would again reveal the allowable substitutions at 2' positions.
  • a logical extension of this analysis is a situation in which one or a few nucleotides of the polymeric ligand is used as a site for chemical derivative exploration.
  • the rest of the ligand serves to anchor in place this monomer (or monomers) on which a variety of derivatives are tested for non-interference with binding and for enhanced affinity.
  • Such explorations may result in small molecules that mimic the structure of the initial ligand framework, and have significant and specific affinity for the target molecule independent of that nucleic acid framework.
  • Such derivatized subunits which may have advantages with respect to mass production, therapeutic routes of administration, delivery, clearance or degradation than the initial SELEX ligand, may become the therapeutic and may retain very little of the original ligand. This approach is thus an additional utility of SELEX.
  • SELEX ligands can allow directed chemical exploration of a defined site on the target molecule known to be important for the target function.
  • This invention includes nucleic acid ligands wherein certain chemical modifications have been made in order to increase the in vivo stability of the ligand or to enhance or mediate the delivery of the ligand.
  • modifications include chemical substitutions at the ribose and/or phosphate positions of a given RNA sequence. See, e.g., Cook, et al. PCT Application WO 9203568; U.S. Pat. No. 5,118,672 of Schinazi et al.; Hobbs et al. Biochem 12:5138 (1973); Guschlbauer et al. Nucleic Acids Res. 4:1933 (1977); Shibahara et al. Nucl. Acids. Res. 15:4403 (1987); Pieken et al. Science 253:314 (1991), each of which is specifically incorporated herein by reference.
  • the walking experiment involves two SELEX experiments performed sequentially.
  • a new candidate mixture is produced in which each of the members of the candidate mixture has a fixed nucleic acid region that corresponds to a SELEX-derived nucleic acid ligand.
  • Each member of the candidate mixture also contains a randomized region of sequences. According to this method it is possible to identify what are referred to as "extended" nucleic acid ligands, that contain regions that may bind to more than one binding domain of a target.
  • the present invention includes computer modeling methods for determining structure of nucleic acid ligands.
  • Secondary structure prediction is a useful guide to correct sequence alignment. It is also a highly useful stepping-stone to correct 3D structure prediction, by constraining a number of bases into A-form helical geometry.
  • thermodynamic rules are inherently inaccurate, typically to 10% or so, and there are many different possible structures lying within 10% of the global energy minimum.
  • the brute force predictive methods is a dotplot: make an N by N plot of the sequence against itself, and mark an X everywhere a basepair is possible. Diagonal runs of X's mark the location of possible helices. Exhaustive tree-searching methods can then search for all possible arrangements of compatible (i.e., non-overlapping) helices of length L or more; energy calculations may be done for these structures to rank them as more or less likely.
  • the advantages of this method are that all possible topologies, including pseudoknotted conformations, may be examined, and that a number of suboptimal structures are automatically generated as well.
  • the disadvantages of the method are that it can run in the worst cases in time proportional to an exponential factor of the sequence size, and may not (depending on the size of the sequence and the actual tree search method employed) look deep enough to find a global minimum.
  • the elegant predictive method, and currently the most used, is the Zuker program.
  • Zuker (1989) Science 244:48-52 Originally based on an algorithm developed by Ruth Nussinov, the Zuker program makes a major simplifying assumption that no pseudoknotted conformations will be allowed. This permits the use of a dynamic programming approach which runs in time proportional to only N 3 to N 4 , where N is the length of the sequence.
  • the Zuker program is the only program capable of rigorously dealing with sequences of than a few hundred nucleotides, so it has come to be the most commonly used by biologists.
  • the central element of the comparative sequence analysis of the present invention is sequence covariations.
  • a covariation is when the identity of one position depends on the identity of another position; for instance, a required Watson-Crick base pair shows strong covariation in that knowledge of one of the two positions gives absolute knowledge of the identity at the other position.
  • Covariation analysis has been used previously to predict the secondary structure of RNAs for which a number of related sequences sharing a common structure exist, such as tRNA, rRNAs, and group I introns. It is now apparent that covariation analysis can be used to detect tertiary contacts as well.
  • M(x,y) The amount of deviation from expectation may be quantified with an information measure M(x,y), the mutual information of x and y: ##EQU1##
  • M(x,y) can be described as the number of bits of information one learns about the identity of position y from knowing just the identity of position y from knowing just the identity of position x. If there is no covariation, M(x,y) is zero; larger values of M(x,y) indicate strong covariation.
  • These covariation values may be used to develop three-dimensional structural predictions.
  • the problem is similar to that of structure determination by NMR. Unlike crystallography, which in the end yields an actual electron density map, NMR yields a set of interatomic distances. Depending on the number of interatomic distances one can get, there may be one, few, or many 3D structures with which they are consistent. Mathematical techniques had to be developed to transform a matrix of interatomic distances into a structure in 3D space. The two main techniques in use are distance geometry and restrained molecular dynamics.
  • the interatomic distances are considered to be coordinates in an N-dimensional space, where N is the number of atoms.
  • N is the number of atoms.
  • the "position" of an atom is specified by N distances to all the other atoms, instead of the three (x,y,z) that we are used to thinking about.
  • Interatomic distances between every atom are recorded in an N by N distance matrix.
  • a complete and precise distance matrix is easily transformed into a 3 by N Cartesian coordinates, using matrix algebra operations.
  • the trick of distance geometry as applied to NMR is dealing with incomplete (only some of the interatomic distances are known) and imprecise data (distances are known to a precision of only a few angstroms at best).
  • Restrained molecular dynamics is a more ad hoc procedure. Given an empirical force field that attempts to describe the forces that all the atoms feel (van der Waals, covalent bonding lengths and angles, electrostatics), one can simulate a number of femtosecond time steps of a molecule's motion, by assigning every atom at a random velocity (from the Boltzmann distribution at a given temperature) and calculating each atom's motion for a femtosecond using Newtonian dynamical equations; that is "molecular dynamics". In restrained molecular dynamics, one assigns extra ad hoc forces to the atoms when they violate specified distance bounds.
  • the covariation values may be transformed into artificial restraining forces between certain atoms for certain distance bounds; varying the magnitude of the force according to the magnitude of the covariance.
  • NMR and covariance analysis generates distance restraints between atoms or positions, which are readily transformed into structures through distance geometry or restrained molecular dynamics.
  • Another source of experimental data which may be utilized to determine the three dimensional structures of nucleic acids is chemical and enzymatic protection experiments, which generate solvent accessibility restraints for individual atoms or positions.
  • nucleic acid ligand for HIV-1 reverse transcriptase HIV-1 reverse transcriptase
  • U.S. patent application Ser. No. 07/714,131 describes the results obtained when SELEX was performed with the HIV-RT target. Inspection of the nucleic acid sequences that were found to have a high affinity to HIV-RT, it was concluded that the nucleic acid ligand solution was configured as a pseudoknot.
  • Described herein are experiments which establish the minimum number of sequences necessary to represent the nucleic acid ligand solution via boundary studies. Also described are the construction of variants of the ligand solution which are used to evaluate the contributions of individual nucleotides in the solution to the binding of the ligand solution to HIV-RT. Also described is the chemical modification of the ligand solution; 1) to corroborate its predicted pseudoknot structure; 2) to determine which modifiable groups are protected from chemical attack when bound to HIV-RT (or become unprotected during binding); and 3) to determine what modifications interfere with binding to HIV-RT (presumably by modification of the three dimensional structure of the ligand solution) and, therefore, which are presumably involved in the proximal contacts with the target.
  • the nucleic acid ligand solution previously determined is shown in FIG. 1. Depicted is an RNA pseudoknot in which Stem 1 (as labeled) is conserved and Stem 2 is relatively non-conserved; X indicates no conservation and X' base-pairs to X.
  • U1 was preferred (existing at this relative position in 11 of the 18 sequences that contributed to the consensus), but A1 was also found frequently (in 6 of the 18).
  • C-G was substituted for the base-pair of G4-C13 and one A-U substitution.
  • the preferred number of nucleotides connecting the two strands of Stem 1 was eight (in 8 of 18).
  • the number and pattern of base-paired nucleotides comprising Stem 2 and the preference for A5 and A12 were derived from the consensus of a secondary SELEX in which the random region was constructed as follows NNUUCCGNNNNNNNNCGGGAAAANNNN (SEQ ID NO:3) (Ns are randomized).
  • NNUUCCGNNNNNNNNCGGGAAAANNNN SEQ ID NO:3
  • One of the ligands was found to significantly inhibit HIV-RT and failed to inhibit AMV or MMLV reverse transcriptases.
  • the first two SELEX experiments in which 32 nucleotide positions were randomized provided high affinity ligands in which there was variable length for Stem 1 at its 5' end; that is, some ligands had the sequence UUCCG which could base pair to CGGGA, UCCG to CGGG or CCG to CGG.
  • RNA's transcribed from oligonucleotide templates were all the same as the complete sequence shown in the upper right hand corner of the figure, except for the varying 5' ends as shown in the boxes A-E lining the left margin.
  • the result is that one 5' U is sufficient for the highest-affinity binding to HIV-RT (boxes A and B), that with no U there is reduced binding (box C), and that any further removal of 5' sequences reduces binding to that of non-specific sequences (box D).
  • the design hereafter referred to as ligand B
  • U 1 was used for the rest of the experiments described here.
  • G4-C13 C-G>U-A>A-U>>>>A-C
  • A-U is about 20-fold reduced in affinity compared to G4-C13 and A-C is at least 100- fold reduced.
  • the base-pairs of Stem 1 seem to exhibit a gradation of resistance to modification such that G4-C13>C3-G14>C2-G15>U1-G16 where G4-C13 is completely resistant to chemical modification and U1-G16 is highly reactive. This suggests that this small helix of the pseudoknot undergoes transient and directional denaturation or "fraying".
  • Binding of protein changes the fraying character of Helix I as shown in FIG. 5 either by stabilizing or protecting it.
  • the natively reactive U1 is also protected upon binding.
  • Binding of protein increases the sensitivity of the base-pair A6-U26 suggesting that this is unpaired in the bound state. This may be an indication of insufficient length of a single nucleotide Loop I during binding, either because it cannot bridge the bound Stem 1 to the end of Stem 2 in the native pseudoknot recognized by RT or because binding increases the length requirement of Loop I by changing the conformation from the native state.
  • A17 and A19 of Loop II are also protected by binding to HIV-RT.
  • the single base bridge A12 is protected upon binding.
  • RNA ligand B was partially modified (with all of the chemicals mentioned above for structure determination). This modified population was bound with varying concentrations of the protein, and the bound species were assayed for the modified positions. From this, it can be determined where modification interferes with binding, and where there is no or little effect.
  • FIG. 6 A schematic diagram summarizing these modification interference results is shown in FIG. 6. As shown, most of the significant interference with binding is clustered on the left hand side of the pseudoknot which contains the Stem 1 and Loop 2. This is also the part of the molecule that was highly conserved (primary sequence) in the collection of sequences isolated by SELEX and where substitution experiments produced the most drastic reduction in binding affinity to HIV-RT.
  • RNA molecules in which there is a 2'-methoxy bonded to the 2' carbon of the ribose instead of the normal hydroxyl group are resistant to enzymatic and chemical degradation.
  • RNA molecules in which there is a 2'-methoxy bonded to the 2' carbon of the ribose instead of the normal hydroxyl group are resistant to enzymatic and chemical degradation.
  • four oligos were prepared as shown in FIG. 7. Because fully substituted 2'-methoxy ligand binds poorly (ligand D), and because we had found that most of the modification interference sites were clustered at one end of the pseudoknot, subsequent attempts to substitute were confined to the non-specific 3' helix as shown in boxes B and C.
  • Oligonucleotides were then prepared in which the allowed substitutions at the ribose of Stem 2 were all 2'-methoxy as in C of FIG. 7 and at the remaining 14 positions mixed synthesis were done with 2'-methoxy and 2'-OH phosphoramidite reagents. These oligos were subjected to selection by HIV-RT followed by alkaline hydrolysis of selected RNAs and gel separation (2'-methoxys do not participate in alkaline hydrolysis as do 2'-hydroxyls). As judged by visual inspection of films (see FIG.
  • U1 can be replaced with A with little loss in affinity but not by C or G. Although U1 probably makes transient base-pairing to G16, modification of U1-N3 with CMCT does not interfere with binding to HIV-RT. However, binding by HIV-RT protects the N3 of U1 perhaps by steric or electrostatic shielding of this position. Substitution with C which forms a more stable base-pair with G16 reduces affinity. Replacement of G16 with A which forms a stable U1-A16 pair abolishes specific affinity for HIV-RT and modification of G16-N1 strongly interferes with binding to HIV-RT. This modification of G16-N1 must prevent a crucial contact with the protein. Why G substitutions for U1 reduce affinity and A substitutions do not is not clear.
  • the G substitution is in a context in which the 5' end of the RNA is one nucleotide shorter, however synthetic RNAs in which U1 is the 5' terminal nucleotide bind with unchanged affinity from those in vitro transcripts with two extra Gs at the 5' end (FIG. 7). Perhaps A at U1 replaces a potential U interaction with a similar or different interaction with HIV-RT a replacement that cannot be performed by C or G at this position.
  • the next base-pair of Stem 1 (C2-G15) cannot be replaced by a G-C base-pair without complete loss of specific affinity for HIV-RT. Modification of the base-pairing faces of either nucleotide strongly interferes with binding to HIV-RT and binding with HIV-RT protects from these modifications. Substitution of the next base-pair, C3-G14, with a G-C pair shows less drastic reduction of affinity, but modification is strongly interfering at this position. Substitution of a C-G pair for G4-C13 has no effect on binding, and substitution of the less stable A-U and U-A pairs allow some specific affinity. Substitution of the non-pairing A-C for these positions abolishes specific binding. This correlates with the appearance of C-G substitutions and one A-U substitution in the original SELEX phylogeny at this position, the non-reactivity of this base-pair in the native state, and the high degree of modification interference found for these bases.
  • Stem 1 the essential components of Stem 1 are a single-stranded 5' nucleotide (U or A) which may make sequence specific contact with the protein and a three base-pair helix (C2-G15, C3-G14, G4-C13) where there are sequence-specific interactions with the HIV-RT at the first two base-pairs and a preference for a strong base-pair (i.e. either C-G or G-C) at the third loop closing position of G4-C13.
  • Loop 2 should be more broadly described as GAXAA (16-20) due to the single-stranded character of G16 which probably interacts with HIV-RT in a sequence-specific manner, as likely do A17 and A19.
  • Stem 2 varies considerably in the pattern and number of base-pairing nucleotides, but from 3' deletion experiments reported here one could hypothesize that a minimum of 3 base-pairs in Stem 2 are required for maximal affinity.
  • At least 2 nucleotides are required in Loop 1 of the bound ligand.
  • FIG. 11 The revised ligand description for HIV-RT obtained based on the methods of this invention is shown in FIG. 11.
  • the major differences between that shown in FIG. 1 is the length of Stem 2, the more degenerate specification of the base-pair G4-C13, the size of Loop 1 (which is directly related to the size of Stem 2) and the single-stranded character of U1 and G16.
  • the SELEX strategy requires 5' and 3' fixed sequences for replication.
  • additional sequences increase the potential for other conformations that compete with that of the high-affinity ligand.
  • additional structural elements that do not directly contribute to affinity such as a lengthened Stem 2 may be selected.
  • G4-C13 Reier et al., Proc. Natl. Acad. Sci. USA, 83:9373-9377 (1986)
  • U1 and G16 may be coincidental to their ability to base-pair; in other nucleic acid ligand-protein complexes such as Klenow fragment/primer-template junction and tRNA/tRNA synthetase there is significant local denaturation of base-paired nucleotides (Freemont et al., Proc. Natl. Acad. Sci. USA, 85:8924 (1988); Ronald et al., Science, 246:1135 (1989)) which may also occur in this case.
  • the distance between the RNase H and polymerase catalytic domains of HIV-RT has recently been determined to be on the order of 18 base-pairs of an A-form RNA-DNA hybrid docked (by computer) in the pocket of a 3.5 ⁇ resolution structure derived from X-ray crystallography (Kohlstaedt et al. Science, 256:1783-1790, (1992)).
  • the distance from the cluster of bases determined to be crucial to this interaction in the pseudoknot and the conserved bases in the extended ligand sequence is approximately 18 base-pairs as well.
  • the pseudoknot interacts with the polymerase catalytic site--in that the ligand has been shown to bind HIV-RT deleted for the RNAse H domain--and that the evolved extension to the pseudoknot may interact with the RNAse H domain.
  • the ligands tested from each of these motifs increase affinity of the ligand B sequence to HIV-RT by at least 10-fold.
  • ligand 6a Described herein are chemical modification experiments performed on ligand 6a designed to confirm the proposed secondary structure, find where binding of the Rev protein protects the ligand from chemical attack, and detect the nucleotides essential for Rev interaction.
  • a secondary SELEX experiment was conducted with biased randomization of the 6a ligand sequence so as to more comprehensively describe a consensus for the highest affinity binding to the HIV-1 Rev protein.
  • Rev ligand 6a Chemical modification studies of the Rev ligand 6a were undertaken to determine its possible secondary structural elements, to find which modifications interfere with the binding of the ligand by Rev, to identify which positions are protected from modification upon protein binding, and to detect possible changes in ligand structure that occur upon binding.
  • the modifying chemicals include ethylnitrosourea (ENU) which modifies phosphates, dimethyl sulfate (DMS) which modifies the base-pairing positions N3 of C and N1 of adenine, kethoxal which modifies base-pairing positions N1 and N2 of guanine, carbodiimide (CMCT) which modifies base-paring position N3 of uracil and to a smaller extent the N1 position of guanine, and diethylpyrocarbonate (DEPC) which modifies the N7 position of adenine and to some extent also the N7 of guanine.
  • ENU modification was assayed by modification-dependent hydrolysis of a labeled RNA chain, while all other modifying agents were used on an extended RNA ligand, with modified positions revealed by primer extension of an annealed oligonucleotide.
  • the chemical probing of the Rev ligand native structure is summarized in FIG. 13.
  • the computer predicted secondary structure Zuker (1989), Science 244:48-52; Jaeger et al. (1989), Proc. Natl. Acad. Sci. USA 86:7706-7710 and native modification data are in general agreement; the ligand is composed of three helical regions, one four-base hairpin loop, and three "bulge” regions (see FIG. 13 for a definition of these structural "elements").
  • U 8 -U 9 are fully modified by CMCT, possibly indicating base orientations into solvent
  • a 13 , A 14 and A 15 are all modified by DMS and DEPC with the strongest modifications occurring on the central A 14 .
  • the bulge opposite to the A 13 -A 15 region shows complete protection of G 26 and very slight modification of A 27 by DMS.
  • Rev-binding RNAs Bartel et al.
  • Modification interference of Rev binding The results of the modification interference studies is summarized in FIG. 14 (quantitative data on individual modifying agents is presented in FIGS. 15 through 19).
  • phosphate and base modification binding interference is clustered into two regions of the RNA ligand. To a first approximation, these regions correspond to two separate motifs present in the SELEX experiments that preceded this present study.
  • Phosphate modification interference is probably the most suggestive of actual sites for ligand-protein contacts, and constitutes an additional criterion for the grouping of the modification interference data into regions.
  • the first region is centered on U 24 -G 25 -G 26 , and includes interference due to phosphate, base-pairing face, and N7 modifications. These same three nucleotides, conserved in the wild-type RRE, were also found to be critical for Rev binding in a modification interference study using short RNAs containing the RRE IIB stem loop (Kjems et al. (1992) EMBO J. 11(3):1119-1129).
  • the second region centers around G 10 -A 11 -G 12 with interference again from phosphate, base-pairing face, and N7 modifications. Additionally, there is a smaller "mini-region" encompassing the stretch C 6 -A 7 -U 8 , with phosphate and base-pairing face modifications interfering with binding.
  • Six rounds of SELEX yielded the list of sequences shown in FIG. 21.
  • the frequency distribution of the nucleotides and base pairs found at each position as it differs from that expected from the input distribution during template synthesis is shown in FIGS. 22 and 23.
  • a new consensus based on these data is shown in FIG. 24.
  • Rev ligand 6a The most significant differences from the sequence of Rev ligand 6a are replacement of the relatively weak base pair A7-U31 with a G-C pair and allowed or prefered substitution of U9 with C, A14 with U, U22 with G.
  • absolutely conserved positions are at sites G10, A11, G12; A15, C16, A17; U24, G25; and C28, U29, C30.
  • No bases were found substituted for G26 and A25, although there was one and three deletions found at those positions respectively.
  • Two labeled transcripts were synthesized, one with a simple ligand 6a-like sequence, and one with substitutions by the significant preferences found in FIG. 24. These RNAs bound identically to Rev protein.
  • SELEX a relatively concentrated pool of heterogeneous RNA sequences (flanked by the requisite fixed sequences) are bound to the protein. In binding assays low concentrations of homogeneous RNA sequence are bound. In SELEX there may be selection for more discriminating conformational certainty due to the increased probability of intermolecular and intramolecular contacts with other RNA sequences. In the therapeutic delivery of concentrated doses of RNA ligands and their modified homologs, these preferences found in secondary SELEXes may be relevant.
  • RNA synthesis In vitro transcription with oligonucleotide templates was conducted as described by Milligan et al. (1988). All synthetic nucleic acids were made on an Applied Biosystems model 394-08 DNA/RNA synthesizer using standard protocols. Deoxyribonucleotide phosphoramidites and DNA synthesis solvents and reagents were purchased from Applied Biosystems. Ribonucleotide and 2'-methoxy-ribonucleotide phosphoramidites were purchased from Glen Research Corporation. For mixed base positions, 0.1M phosphoramidite solutions were mixed by volume to the proportions indicated. Base deprotection was carried out at 55° C. for 6 hours in 3:1 ammonium hydroxide:ethanol.
  • t-butyl-dimethylsilyl protecting groups were removed from the 2'-OH groups of synthetic RNAs by overnight treatment in tetrabutylammonium fluoride. The deprotected RNAs were then phenol extracted, ethanol precipitated and purified by gel electrophoresis.
  • RNAs for refinement of the 5' and 3' boundaries and for determination of the effect of substitutions were labeled during transcription with T7 RNA polymerase as described in Tuerk et al. (1990) except that a-32P-ATP was used, in reactions of 0.5 mM C,G, and UTP with 0.05 mM ATP.
  • Synthetic oligonucleotides and phosphatased transcripts were kinased as described in Gauss et al. (1987).
  • RNA-protein binding reactions were done in a "binding buffer" of 200 mM KOAc, 50 mM Tris-HCl pH 7.7, 10 mM dithiothreitol with exceptions noted for chemical protection experiments below.
  • RNA and protein dilutions were mixed and stored on ice for 30 minutes then transferred to 37° C. for 5 minutes.
  • binding assays the reaction volume was 60 ul of which 50 ul was assayed.
  • Each reaction was suctioned through a pre-wet (with binding buffer) nitrocellulose filter and rinsed with 3 mls of binding buffer after which it was dried and counted for assays or subjected to elution and assayed for chemical modification.
  • results were plotted and the protein concentration at which half-maximal binding occurred (the approximate Kd in conditions of protein excess) was determined graphically.
  • RNAs that were modified under denaturing conditions were selected at concentrations of 20, 4 and 0.8 nanomolar HIV-RT (in volumes of 1, 5 and 25 mls of binding buffer.) The amount of RNA added to each reaction was equivalent for each experiment (approximately 1-5 picomoles). RNA was eluted from filters as described in Tuerk et al. (1990) and assayed for modified positions.
  • ENU ethylnitrosourea
  • DMS dimethyl sulfate
  • CMCT carbodiimide
  • DEPC diethyl pyrocarbonate
  • concentrations of modifying chemical reagent were identical for denaturing conditions (except where noted for DEPC); those conditions were 7M urea, 50 mM Tris-HCl pH 7.7, 1 mM EDTA at 90° C. for 1-5 minutes except during modification with ENU which was done in the absence of 7M urea.
  • RNA in the presence of HIV-RT was modified. Conditions were as for modification of native RNA. Concentrations of HIV-RT were approximately 10-fold excess over RNA concentration. In general protein concentrations ranged from 50 nM to 1 uM.
  • RNA was transcribed from PCRd templates synthesized from the following oligonucleotides:
  • SELEX was performed as described previously with HIV-RT with the following exceptions.
  • concentration of HIV-RT in the binding reaction of the first SELEX round was 13 nanomolar
  • RNA at 10 micromolar, in 4 mls of binding buffer in the rounds 2 through 9 selection was done with 2.6 nanomolar HIV-RT, 1.8 micromolar RNA in 20 mls of buffer, in rounds 10-14 we used 1 nanomolar HIV-RT, 0.7 micromolar RNA in 50 mls, and for rounds 15 and 16 we used 0.5 nanomolar HIV-RT, 0.7 micromolar RNA in 50 mls of binding buffer.
  • RNA pseudoknots that inhibit HIV-1 reverse transcriptase Proc. Natl. Acad. Sci. USA 89:6988-6992.
  • EXAMPLE II ELUCIDATION OF IMPROVED NUCLEIC ACID LIGAND SOLUTIONS FOR HIV-1 REV PROTEIN
  • RNA for modification was obtained from T7 RNA polymerase transcription of synthetic oligonucleotide templates.
  • ENU modification was carried out on the ligand sequence as shown in FIG. 12.
  • DMS, kethoxal, CMCT, and DEPC modifications were carried out on a extended ligand sequence, and analyzed by reverse transcription with the synthetic oligonucleotide primer shown in FIG. 12.
  • RNA under native conditions was performed in 200 mM KOAc, 50 mM Tris-HCl pH 7.7, 1 mM EDTA at 37 ° C.
  • Modification under denaturing conditions was done in 7M urea, 50 mM Tris-HCl pH 7.7 at 90 ° C.
  • Concentration of modifying agents and incubation times are as follows: ethylnitrosourea (ENU)-1/5 dilution v/v of ethanol saturated with ENU, native 1-3 hours, denaturing 5 minutes; dimethyl sulfate (DMS)-1/750-fold dilution v/v, native 8 minutes, denaturing 1 minute; kethoxal--0.5 mg/ml, native 5 minutes, denaturing 2 minutes; carbodiimide (CMCT)--10 mg/ml, native 30 minutes, denaturing 3 minutes; diethyl pyrocarbonate (DEPC)--1/10 dilution v/v, native 10 minutes, denaturing 1 minute.
  • ENU ethylnitrosourea
  • DMS dimethyl sulfate
  • CMCT carbodiimide
  • DEPC diethyl pyrocarbonate
  • RNAs chemically modified under denaturing conditions were selected for Rev binding through filter partitioning. Selections were carried out at Rev concentrations of 30, 6, and 1.2 nanomolar (in respective volumes of 1, 5, and 25 mls of binding buffer; 200 mM KOAc, 50 mM Tris-HCl pH 7.7, and 10 mM dithiothreitol), Approximately 3 picomoles of modified RNA were added to each protein solution, mixed and stored on ice for 15 minutes, and then transferred to 37° C. for 10 minutes. Binding solutions were passed through pre-wet nitrocellulose filters, and rinsed with 5 mls of binding buffer. RNA was eluted from the filters as described in Tuerk et al. (1990) Science 24:505-510 and assayed for modified positions that remained. Modified RNA was also spotted on filters and eluted to check for uniform recovery of modified RNA.
  • the extent of modification interference was determined by densitometric scanning of autoradiographs using LKB (ENU) and Molecular Dynamics (DMS, kethoxal, CMCT, and DEPC) laser densitometers. Values for modified phosphates and bases were normalized to a chosen modified position for both selected and unselected lanes; the values for the modified positions in the selected lane were then divided by the corresponding positions in the unselected lane (for specific normalizing positions see FIGS. 15-19). Values above 4.0 for modified bases and phosphates are designated as strongly interfering, and values above 2.0 are termed slightly interfering.
  • “Footprinting" of the Rev ligand, modification of the RNA ligand in the presence of Rev protein was performed in 200 mM KOAc, 50 mM Tris-Cl pH 7.7, 1 mM DTT, and 5 mM MgCl. Concentration of protein was 500 nanomolar, and approximately in 3-fold molar excess over RNA concentration. Modification with protein present was attempted with all modifying agents listed above except ethylnitrosourea (ENU).
  • ENU ethylnitrosourea
  • the templates for in vitro transcription were prepared by PCR from the following oligonucleotides:
  • the small case letters in the template oligo indicates that at each position that a mixture of reagents were used in synthesis by an amount of 62.5% of the small case letter, and 12.5% each of the other three nucleotides.
  • HIV-1 Rev protein was purchased from American Biotechnologies, Inc.

Abstract

Methods for the identification and production of improved nucleic acid ligands are based on the SELEX process. Nucleic acid ligands to HIV-RT and HIV-1 Rev are identified according to the methods described herein.

Description

This application is a Continuation-in-Part of U.S. patent application Ser. No. 07/714,131, filed Jun. 10, 1991, entitled Nucleic Acid Ligands and U.S. patent application Ser. No. 07/536,428, filed Jun. 11, 1990, entitled Systematic Evolution of Ligands by Exponential Enrichment now abandoned.
FIELD OF THE INVENTION
Described herein are methods for identifying and producing nucleic acid ligands. Nucleic acid ligands are double or single stranded DNA or RNA species that bind specifically to a desired target molecule. The basis for identifying nucleic acid ligands is a method that is called SELEX, an acronym for Systematic Evolution of Ligands by Exponential enrichment. The methods of the present invention include means for analyzing and applying the information learned from the SELEX method to create an improved nucleic acid ligand for the selected target. These methods include computer modeling, boundary determination methods and chemical modification methods. According to the methods of this invention it is possible to determine: 1) which nucleic acid residues of a nucleic acid ligand are critical in binding to the selected target; 2) which nucleic acid residues affect the structural conformation of the nucleic acid ligand; and 3) what is the three-dimensional structure of the nucleic acid ligand. This information allows for the identification and production of improved nucleic acid ligands that have superior binding capacity to the target as well as enhanced structural stability. This information may also be utilized to produce non-nucleic acid or hybrid-nucleic acid species that also function as ligands to the target. The methods of the present invention further provide an analysis of the target species that can be used in the preparation of therapeutic and/or diagnostic methods.
BACKGROUND OF THE INVENTION
Most proteins or small molecules are not known to specifically bind to nucleic acids. The known protein exceptions are those regulatory proteins such as repressors, polymerases, activators and the like which function in a living cell to bring about the transfer of genetic information encoded in the nucleic acids into cellular structures and the replication of the genetic material. Furthermore, small molecules such as GTP bind to some intron RNAs.
Living matter has evolved to limit the function of nucleic acids to a largely informational role. The Central Dogma, as postulated by Crick, both originally and in expanded form, proposes that nucleic acids (either RNA or DNA) can serve as templates for the synthesis of other nucleic acids through replicative processes that "read" the information in a template nucleic acid and thus yield complementary nucleic acids. All of the experimental paradigms for genetics and gene expression depend on these properties of nucleic acids: in essence, double-stranded nucleic acids are informationally redundant because of the chemical concept of base pairs and because replicative processes are able to use that base pairing in a relatively error-free manner.
The individual components of proteins, the twenty natural amino acids, possess sufficient chemical differences and activities to provide an enormous breadth of activities for both binding and catalysis. Nucleic acids, however, are thought to have narrower chemical possibilities than proteins, but to have an informational role that allows genetic information to be passed from virus to virus, cell to cell, and organism to organism. In this context nucleic acid components, the nucleotides, must possess only pairs of surfaces that allow informational redundancy within a Watson-Crick base pair. Nucleic acid components need not possess chemical differences and activities sufficient for either a wide range of binding or catalysis.
However, some nucleic acids found in nature do participate in binding to certain target molecules and even a few instances of catalysis have been reported. The range of activities of this kind is narrow compared to proteins and more specifically antibodies. For example, where nucleic acids are known to bind to some protein targets with high affinity and specificity, the binding depends on the exact sequences of nucleotides that comprise the DNA or RNA ligand. Thus, short double-stranded DNA sequences are known to bind to target proteins that repress or activate transcription in both prokaryotes and eukaryotes. Other short double-stranded DNA sequences are known to bind to restriction endonucleases, protein targets that can be selected with high affinity and specificity. Other short DNA sequences serve as centromeres and telomeres on chromosomes, presumably by creating ligands for the binding of specific proteins that participate in chromosome mechanics. Thus, double-stranded DNA has a well-known capacity to bind within the nooks and crannies of target proteins whose functions are directed to DNA binding. Single-stranded DNA can also bind to some proteins with high affinity and specificity, although the number of examples is rather smaller. From the known examples of double-stranded DNA binding proteins, it has become possible to describe the binding interactions as involving various protein motifs projecting amino acid side chains into the major groove of B form double-stranded DNA, providing the sequence inspection that allows specificity.
Double-stranded RNA occasionally serves as a ligand for certain proteins, for example, the endonuclease RNase III from E. coli. There are more known instances of target proteins that bind to single-stranded RNA ligands, although in these cases the single-stranded RNA often forms a complex three-dimensional shape that includes local regions of intramolecular double-strandedness. The amino-acyl tRNA synthetases bind tightly to tRNA molecules with high specificity. A short region within the genomes of RNA viruses binds tightly and with high specificity to the viral coat proteins. A short sequence of RNA binds to the bacteriophage T4-encoded DNA polymerase, again with high affinity and specificity. Thus, it is possible to find RNA and DNA ligands, either double- or single-stranded, serving as binding partners for specific protein targets. Most known DNA binding proteins bind specifically to double-stranded DNA, while most RNA binding proteins recognize single-stranded RNA. This statistical bias in the literature no doubt reflects the present biosphere's statistical predisposition to use DNA as a double-stranded genome and RNA as a single-stranded entity in the roles RNA plays beyond serving as a genome. Chemically there is no strong reason to dismiss single-stranded DNA as a fully able partner for specific protein interactions.
RNA and DNA have also been found to bind to smaller target molecules. Double-stranded DNA binds to various antibiotics, such as actinomycin D. A specific single-stranded RNA binds to the antibiotic thiostreptone; specific RNA sequences and structures probably bind to certain other antibiotics, especially those whose functions is to inactivate ribosomes in a target organism. A family of evolutionary related RNAs binds with specificity and decent affinity to nucleotides and nucleosides (Bass, B. and Cech, T. (1984) Nature 308:820-826) as well as to one of the twenty amino acids (Yarus, M. (1988) Science 240:1751-1758). Catalytic RNAs are now known as well, although these molecules perform over a narrow range of chemical possibilities, which are thus far related largely to phosphodiester transfer reactions and hydrolysis of nucleic acids.
Despite these known instances, the great majority of proteins and other cellular components are thought not to bind to nucleic acids under physiological conditions and such binding as may be observed is non-specific. Either the capacity of nucleic acids to bind other compounds is limited to the relatively few instances enumerated supra, or the chemical repertoire of the nucleic acids for specific binding is avoided (selected against) in the structures that occur naturally. The present invention is premised on the inventors' fundamental insight that nucleic acids as chemical compounds can form a virtually limitless array of shapes, sizes and configurations, and are capable of a far broader repertoire of binding and catalytic functions than those displayed in biological systems.
The chemical interactions have been explored in cases of certain known instances of protein-nucleic acid binding. For example, the size and sequence of the RNA site of bacteriophage R17 coat protein binding has been identified by Uhlenbeck and coworkers. The minimal natural RNA binding site (21 bases long) for the R17 coat protein was determined by subjecting variable-sized labeled fragments of the mRNA to nitrocellulose filter binding assays in which protein-RNA fragment complexes remain bound to the filter (Carey et al. (1983) Biochemistry 22:2601). A number of sequence variants of the minimal R17 coat protein binding site were created in vitro in order to determine the contributions of individual nucleic acids to protein binding (Uhlenbeck et al. (1983) J. Biomol. Structure Dynamics 1:539 and Romaniuk et al. (1987) Biochemistry 26:1563). It was found that the maintenance of the hairpin loop structure of the binding site was essential for protein binding but, in addition, that nucleotide substitutions at most of the single-stranded residues in the binding site, including a bulged nucleotide in the hairpin stem, significantly affected binding. In similar studies, the binding of bacteriophage Qβ coat protein to its translational operator was examined (Witherell and Uhlenbeck (1989) Biochemistry 28:71). The Qβ coat protein RNA binding site was found to be similar to that of R17 in size, and in predicted secondary structure, in that it comprised about 20 bases with an 8 base pair hairpin structure which included a bulged nucleotide and a 3 base loop. In contrast to the R17 coat protein binding site, only one of the single-stranded residues of the loop is essential for binding and the presence of the bulged nucleotide is not required. The protein-RNA binding interactions involved in translational regulation display significant specificity.
Nucleic acids are known to form secondary and tertiary structures in solution. The double-stranded forms of DNA include the so-called B double-helical form, Z-DNA and superhelical twists (Rich, A. et al. (1984) Ann. Rev. Biochem. 53:791-846). Single-stranded RNA forms localized regions of secondary structure such as hairpin loops and pseudoknot structures (Schimmel, P. (1989) Cell 58:9-12). However, little is known concerning the effects of unpaired loop nucleotides on stability of loop structure, kinetics of formation and denaturation, thermodynamics, and almost nothing is known of tertiary structures and three dimensional shape, nor of the kinetics and thermodynamics of tertiary folding in nucleic acids (Tuerk, C. et al. (1988) Proc. Natl. Acad. Sci. USA 85.:1364-1368).
A type of in vitro evolution was reported in replication of the RNA bacteriophage Qβ. Mills, D. R. et al. (1967) Proc. Natl. Acad. Sci USA 58:217-224; Levisohn, R. and Spiegeleman, S. (1968) Proc. Natl. Acad. Sci. USA 60:866-872; Levisohn, R. and Spiegelman S. (1969) Proc. Natl. Acad. Sci. USA 63:805-811; Saffhill, R. et al. (1970) J. Mol. Biol. 51:531-539; Kacian, D. L. et al. (1972) Proc. Natl. Acad. Sci. USA 69:3038-3042; Mills, D. R. et al. (1973) Science 180:916-927. The phage RNA serves as a poly-cistronic messenger RNA directing translation of phage-specific proteins and also as a template for its own replication catalyzed by Qβ RNA replicase. This RNA replicase was shown to be highly specific for its own RNA templates. During the course of cycles of replication in vitro small variant RNAs were isolated which were also replicated by Qβ replicase. Minor alterations in the conditions under which cycles of replication were performed were found to result in the accumulation of different RNAs, presumably because their replication was favored under the altered conditions. In these experiments, the selected RNA had to be bound efficiently by the replicase to initiate replication and had to serve as a kinetically favored template during elongation of RNA. Kramer et al. (1974) J. Mol. Biol. 89:719 reported the isolation of a mutant RNA template of Qβ replicase, the replication of which was more resistant to inhibition by ethidium bromide than the natural template. It was suggested that this mutant was not present in the initial RNA population but was generated by sequential mutation during cycles of in vitro replication with Qβ replicase. The only source of variation during selection was the intrinsic error rate during elongation by Qβ replicase. In these studies what was termed "selection" occurred by preferential amplification of one or more of a limited number of spontaneous variants of an initially homogenous RNA sequence. There was no selection of a desired result, only that which was intrinsic to the mode of action of Qβ replicase.
Joyce and Robertson (Joyce (1989) in RNA: Catalysis, Splicing, Evolution, Belfort and Shub (eds.), Elsevier, Amsterdam pp. 83-87; and Robertson and Joyce (1990) Nature 344:467) reported a method for identifying RNAs which specifically cleave single-stranded DNA. The selection for catalytic activity was based on the ability of the ribozyme to catalyze the cleavage of a substrate ssRNA or DNA at a specific position and transfer the 3'-end of the substrate to the 3'-end of the ribozyme. The product of the desired reaction was selected by using an oligodeoxynucleotide primer which could bind only to the completed product across the junction formed by the catalytic reaction and allowed selective reverse transcription of the ribozyme sequence. The selected catalytic sequences were amplified by attachment of the promoter of T7 RNA polymerase to the 3'-end of the cDNA, followed by transcription to RNA. The method was employed to identify from a small number of ribozyme variants the variant that was most reactive for cleavage of a selected substrate.
The prior art has not taught or suggested more than a limited range of chemical functions for nucleic acids in their interactions with other substances: as targets for proteins evolved to bind certain specific oligonucleotide sequences; and more recently, as catalysts with a limited range of activities. Prior "selection" experiments have been limited to a narrow range of variants of a previously described function. Now, for the first time, it will be understood that the nucleic acids are capable of a vastly broad range of functions and the methodology for realizing that capability is disclosed herein.
U.S. patent application Ser. No. 07/536,428 filed Jun. 11, 1990, of Gold and Tuerk, entitled Systematic Evolution of Ligands by Exponential Enrichment, now abandoned and U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991 of Gold and Tuerk, entitled Nucleic Acid Ligands (See also WO 91/19813) describe a fundamentally novel method for making a nucleic acid ligand for any desired target. Each of these applications, collectively referred to herein as the SELEX Patent Applications, is specifically incorporated herein by reference.
The method of the SELEX Patent Applications is based on the unique insight that nucleic acids have sufficient capacity for forming a variety of two- and three-dimensional structures and sufficient chemical versatility available within their monomers to act as ligands (form specific binding pairs) with virtually any chemical compound, whether large or small in size.
The method involves selection from a mixture of candidates and step-wise iterations of structural improvement, using the same general selection theme, to achieve virtually any desired criterion of binding affinity and selectivity. Starting from a mixture of nucleic acids, preferably comprising a segment of randomized sequence, the method, termed SELEX herein, includes steps of contacting the mixture with the target under conditions favorable for binding, partitioning unbound nucleic acids from those nucleic acids which have bound to target molecules, dissociating the nucleic acid-target pairs, amplifying the nucleic acids dissociated from the nucleic acid-target pairs to yield a ligand-enriched mixture of nucleic acids, then reiterating the steps of binding, partitioning, dissociating and amplifying through as many cycles as desired.
While not bound by a theory of preparation, SELEX is based on the inventors' insight that within a nucleic acid mixture containing a large number of possible sequences and structures there is a wide range of binding affinities for a given target. A nucleic acid mixture comprising, for example a 20 nucleotide randomized segment can have 420 candidate possibilities. Those which have the higher affinity constants for the target are most likely to bind. After partitioning, dissociation and amplification, a second nucleic acid mixture is generated, enriched for the higher binding affinity candidates. Additional rounds of selection progressively favor the best ligands until the resulting nucleic acid mixture is predominantly composed of only one or a few sequences. These can then be cloned, sequenced and individually tested for binding affinity as pure ligands.
Cycles of selection and amplification are repeated until a desired goal is achieved. In the most general case, selection/amplification is continued until no significant improvement in binding strength is achieved on repetition of the cycle. The method may be used to sample as many as about 1018 different nucleic acid species. The nucleic acids of the test mixture preferably include a randomized sequence portion as well as conserved sequences necessary for efficient amplification. Nucleic acid sequence variants can be produced in a number of ways including synthesis of randomized nucleic acid sequences and size selection from randomly cleaved cellular nucleic acids. The variable sequence portion may contain fully or partially random sequence; it may also contain subportions of conserved sequence incorporated with randomized sequence. Sequence variation in test nucleic acids can be introduced or increased by mutagenesis before or during the selection/amplification iterations.
In one embodiment of the method of the SELEX Patent Applications, the selection process is so efficient at isolating those nucleic acid ligands that bind most strongly to the selected target, that only one cycle of selection and amplification is required. Such an efficient selection may occur, for example, in a chromatographic-type process wherein the ability of nucleic acids to associate with targets bound on a column operates in such a manner that the column is sufficiently able to allow separation and isolation of the highest affinity nucleic acid ligands.
In many cases, it is not necessarily desirable to perform the iterative steps of SELEX until a single nucleic acid ligand is identified. The target-specific nucleic acid ligand solution may include a family of nucleic acid structures or motifs that have a number of conserved sequences and a number of sequences which can be substituted or added without significantly effecting the affinity of the nucleic acid ligands to the target. By terminating the SELEX process prior to completion, it is possible to determine the sequence of a number of members of the nucleic acid ligand solution family.
A variety of nucleic acid primary, secondary and tertiary structures are known to exist. The structures or motifs that have been shown most commonly to be involved in non-Watson-Crick type interactions are referred to as hairpin loops, symmetric and asymmetric bulges, psuedoknots and myriad combinations of the same. Almost all known cases of such motifs suggest that they can be formed in a nucleic acid sequence of no more than 30 nucleotides. For this reason, it is often preferred that SELEX procedures with contiguous randomized segments be initiated with nucleic acid sequences containing a randomized segment of between about 20-50 nucleotides.
The SELEX Patent Applications also describe methods for obtaining nucleic acid ligands that bind to more than one site on the target molecule, and to nucleic acid ligands that include non-nucleic acid species that bind to specific sites on the target. The SELEX method provides means for isolating and identifying nucleic acid ligands which bind to any envisonable target. However, in preferred embodiments the SELEX method is applied to situations where the target is a protein, including both nucleic acid-binding proteins and proteins not known to bind nucleic acids as part of their biological function.
Little is known about RNA structure at high resolution. The basic A-form helical structure of double stranded RNA is known from fiber diffraction studies. X-ray crystallography has yielded the structure of a few tRNAs and a short poly-AU helix. The X-ray structure of a tRNA/synthetase RNA/protein complex has also been solved. The structures of two tetranucleotide hairpin loops and one model pseudoknot are know from NMR studies.
There are several reasons behind the paucity of structural data. Until the advent of in vitro RNA synthesis, it was difficult to isolate quantities of RNA sufficient for structural work. Until the discovery of catalytic RNAs, there were few RNA molecules considered worthy of structural study. Good tRNA crystals have been difficult to obtain, discouraging other crystal studies. The technology for NMR study of molecules of this size has only recently become available.
As described above, several examples of catalytic RNA structures are known, and the SELEX technology has been developed which selects RNAs that bind tightly to a variety of target molecules--and may eventually be able to select for new catalytic RNA structures as well. It has become important to know the structure of these molecules, in order to learn how exactly they work, and to use this knowledge to improve upon them.
It would be desirable to understand enough about RNA folding to be able to predict the structure of an RNA with less effort than resorting to rigorous NMR, and X-ray crystal structure determination. For both proteins and RNAs, there has always been a desire to be able to compute structures based on sequences, and with limited (or no) experimental data.
Protein structure prediction is notoriously difficult. To a first approximation, the secondary structure and tertiary structure of proteins form cooperatively; protein folding can be approximated thermodynamically by a two-state model, with completely folded and completely unfolded states. This means that the number of degrees of freedom for modeling a protein structure are very large; without predictable intermediates, one cannot break the prediction problem into smaller, manageable sub problems. In contrast, RNAs often appear to make well-defined secondary structures which provide more stability than the tertiary interactions. For example, the tertiary structure of tRNA can be disrupted without disrupting the secondary structure by chelation of magnesium or by raising the temperature. Secondary structure prediction for RNAs is well-understood, and is generally quite accurate for small RNA molecules. For RNAs, structural prediction can be broken into subproblems; first, predict the secondary structure; then, predict how the resulting helices and remaining single strands are arranged relative to each other.
For RNA, the first attempts at structural prediction were for tRNAs. The secondary structure of the canonical tRNA cloverleaf was known from comparative sequence analysis, reducing the problem to one of arranging four short A-form helices in space relative to each other. Manual CPK modeling, back-of-the-envelope energy minimization, and a few distance restraints available from crosslinking studies and phylogenetic covariations were used to generate a tRNA model--which unfortunately proved wrong when the first crystal structure of phenylalanine tRNA was solved a few years later.
Computer modeling has supplanted manual modeling, relieving the model-builder of the difficulties imposed by gravitation and mass. Computer modeling can only be used without additional experimental data for instances in which a homologous structure is known; for instance, the structure of the 3' end of the turnip yellow mosaic virus RNA genome was modeled, based on the known 3D structure of tRNA and the knowledge that the 3' end of TYMV is recognized as tRNA-like by a number of cellular tRNA modification enzymes. This model was the first 3D model of an RNA pseudoknot; the basic structure of an isolated model pseudoknot has been corroborated by NMR data.
Computer modeling protocols have been used, restrained by the manual inspection of chemical and enzymatic protection data, to model the structures of several RNA molecules. In one isolated substructure, one model for the conformation of a GNRA tetranucleotide loop has been shown to be essentially correct by NMR study of an isolated GNRA hairpin loop.
Francois Michel ((1989) Nature 342:391) has constructed a model for the catalytic core of group I introns. Like the tRNAs, the secondary structure of group I intron cores is well-known from comparative sequence analysis, so the problem is reduced to one of properly arranging helices and the remaining single-stranded regions. Michel ((1989) supra) analyzed an aligned set of 87 group I intron sequences by eye and detected seven strong pairwise and triplet covariations outside of the secondary structure, which he interpreted as tertiary contacts and manually incorporated as restraints on his model. As yet, there is no independent confirmation of the Michel model.
Others have attempted to devise an automated procedure to deal with distance restraints from crosslinking, fluorescence transfer, or phylogentic co-variation. The RNA is treated as an assemblage of cylinders (A-form helices) and beads (single-stranded residues), and a mathematical technique called distance geometry is used to generate arrangements of these elements which are consistent with a set of distance restraints. Using a small set of seven distance restraints on the phenylalanine tRNA tertiary structure, this protocol generated the familiar L-form of the tRNA structure about 2/3 of the time.
SUMMARY OF THE INVENTION
The present invention includes methods for identifying and producing nucleic acid ligands and the nucleic acid ligands so identified and produced. The SELEX method described above allows for the identification of a single nucleic acid ligand or a family of nucleic acid ligands to a given target. The methods of the present invention allow for the analysis of the nucleic acid ligand or family of nucleic acid ligands obtained by SELEX in order to identify and produce improved nucleic acid ligands.
Included in this invention are methods for determining the three-dimensional structure of nucleic acid ligands. Such methods include mathematical modeling and structure modifications of the SELEX derived ligands. Further included are methods for determining which nucleic acid residues in a nucleic acid ligand are necessary for maintaining the three-dimensional structure of the ligand, and which residues interact with the target to facilitate the formation of ligand-target binding pairs.
In one embodiment of the present invention, nucleic acid ligands are desired for their ability to inhibit one or more of the biological activities of the target. In such cases, methods are provided for determining whether the nucleic acid ligand effectively inhibits the desired biological activity.
Further included in this invention are methods for identifying tighter-binding RNA ligands and smaller, more stable ligands for use in pharmaceutical or diagnostic purposes.
The present invention includes improved nucleic acid ligands to the HIV-RT and HIV-1 Rev proteins. Also included are nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind HIV-RT or the HIV-1 Rev protein as the nucleic acid ligands specifically identified herein.
Also included within the scope of the invention is a method for performing sequential SELEX experiments in order to identify extended nucleic acid ligands. In particular, extended nucleic acid ligands to the HIV-RT protein are disclosed. Nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind HIV-RT as the extended HIV-RT nucleic acid ligands are also included in this invention.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 depicts the consensus pseudoknot derived from primary and secondary SELEX experiments describing high affinity inhibitory ligands of HIV-1 reverse transcriptase (HIV-RT). The consensus secondary structure is a pseudoknot; the 5' helix of that pseudoknot (Stem 1) is conserved at the primary sequence level and the 3' helix or Stem 2 is not. X indicates a nucleotide position that is non-conserved; X-X' indicates a preferred base-pair. The 26 nucleotide positions are numbered as shown.
FIG. 2A depicts refinement of the 5' information boundary. A set of model ligands was synthesized with T7 RNA polymerase from template oligos. Milligan et al. Nucl. Acid. Res., 15:8783-8798 (1987). Illustrated in the upper left is the complete ligand B. On the right margin are shown the variations in the individual ligands A through E that occur in the boxed areas.
FIG. 2B depicts graphically the :individual binding curves for these model ligands.
FIG. 3 depicts the effect of various nucleotide substitutions within the ligand B sequence on binding to HIV-RT. Illustrated are the various substitutions and resultant affinities to HIV-RT expressed relative to the binding of ligand B. Ligand B was a control tested in each experiment; the affinity of ligand B is normalized as 1.0 and the relative affinity (Kd of ligand B is divided by the Kd of each ligand) is shown. Also shown are the affinities of various truncations of ligand B. The value associated with the asterisked G--G which replaces U1-G16 comes from ligand C of FIG. 2.
FIG. 4 depicts a chemical probe of the native versus denatured conformations of ligand B. The various nucleotides of ligand B were reacted with chemicals under native and denaturing conditions, assayed for the modified positions, electrophoresed and visualized for comparison. indicate highly reactive base-pairing groups of the base at that position and □ partially reactivity; indicates strong reactivity of purine N7 positions and .increment. partial reactivity (to modification with DEPC). The question marks indicate that these positions on G(-2) and G(-1) could not be distinguished due to band crowding on the gel.
FIG. 5 depicts reactivities of modifiable groups of ligand B when bound to HIV-RT. Diagrammed are those groups that show altered reactivity when bound to HIV-RT as compared to that of the native conformation.
FIG. 6 depicts modification interference results for ligand B complexing with HIV-RT. Symbols for modification are as in the boxed legend. The modifications indicated are those that are strongly (filled symbols) or partially (unfilled symbols) selected against by binding to HIV-RT (reflected by decreased modification at those positions in the selected population).
FIG. 7A depicts substitution of 2'-methoxy for hydroxyl on the riboses of the ligand B sequence shown in the upper right. Illustrated in the upper right is the complete ligand B. On the right margin are shown the variations in the individual ligands A through D that occur in ligand B.
FIG. 7B depicts graphically the individual binding curves for ligands A-D.
FIG. 8 depicts selection by HIV-RT from mixed populations of 2'-methoxy ribose versus 2'-hydroxyl at positions U1 through A5 and A12 through A20. An oligonucleotide was synthesized with the following sequence:
5'-(AAAAA).sub.d (UCCGA).sub.x (AGUGCA).sub.m (ACGGGAAAA).sub.x (UGCACU).sub.m-3'
where subscripted "d" indicates 2'-deoxy, subscripted "x" that those nucleotides are mixed 50--50 for phosphoramidite reagents resulting in 2'-methoxy or 2'-hydroxyl on the ribose, and subscripted "m" indicating that those nucleotides are all 2'-methoxy on the ribose.
FIG. 9 shows the starting RNA (SEQ ID NO:37) and the collection of sequences, grouped into two motifs, Extension Motif I (SEQ ID NOS: 14-27) and Extension Motif II (SEQ ID NO8:28-33), obtained from SELEX with HIV-RT as part of a walking experiment.
FIG. 10 illustrates the consensus extended HIV-RT ligand obtained from the list of sequences shown in FIG. 9.
FIG. 10A illustrates the secondary structure of the first 25 bases of the starting material (SEQ ID NO:37) shown in FIG. 9.
FIGS. 10B and 10C illustrate the consensus extended HIV-RT ligands obtained from the list of sequences, Extension Motif I (SEQ ID NOS:14-27) (FIG. 10B) and Extension Motif II (SEQ ID NO8:28-33) (FIG. 10C), shown in FIG. 9.
FIG. 11 illustrates the revised description of the pseudoknot ligand of HIV-RT. In addition to the labeling conventions of FIG. 1, the S-S' indicates the preferred C-G or G-C base-pair at this position.
FIG. 12A shows the sequence of a high-affinity RNA ligand for HIV-1 Rev protein obtained from SELEX experiments. Shown is the numbering scheme used for reference to particular bases in the RNA. This sequence was used for chemical modification with ENU.
FIG. 12B shows the extended RNA sequence used in chemical modification experiments with DMS, kethoxal, CMCT, and DEPC.
FIG. 12C shows the sequence of the oligonucleotide used for primer extension of the extended ligand sequence.
FIG. 13 depicts the results of chemical modification of the HIV-1 Rev ligand RNA under native conditions, a) lists chemical modifying agents, their specificity, and the symbols denoting partial and full modification. The RNA sequence is shown, with degree and type of modification displayed for every modified base, b) depicts the helical, bulge, and hairpin structural elements of the HIV-1 Rev RNA ligand corresponding to the modification and computer structural prediction data.
FIG. 14 depicts the results of chemical modification of the ligand RNA that interferes with binding to the HIV-1 Rev protein. Listed are the modifications which interfere with protein binding, classified into categories of strong interference and slight interference. Symbols denote either base-pairing modifications, N7 modifications, or phosphate modifications.
FIG. 15 depicts the modification interference values for phosphate alkylation. Data is normalized to A17 3' phosphate.
FIG. 16 depicts the modification interference values for DMS modification of N3C and N1A. Data is normalized to C36; A34.
FIG. 17 depicts the modification interference values for kethoxal modification of N1G and N2G. Data is normalized to G5.
FIG. 18 depicts the modification interference values for CMCT modification of N3U and N1G. Data is normalized to U38.
FIG. 19 depicts the modification interference values for DEPC modification of N7A and N7G. Data normalized to G19; A34.
FIG. 20 depicts the chemical modification of the RNA ligand in the presence of the HIV-1 Rev protein. Indicated are those positions that showed either reduced modification or enhanced modification in the presence of protein as compared to modification under native conditions but without protein present.
FIG. 21 shows the 5' and 3' sequences which flank the "6a" biased random region used in SELEX. The template which produced the initial RNA population was constructed from the following oligonucleotides:
5'-CCCGGATCCTCTTTACCTCTGTGTGagatacagagtccacaaacgtgttc tcaatgcacccGGTCGGAAGGCCATCAATAGTCCC-3' (template oligo) (SEQ ID NO: 9)
5'-CCGAAGCTTAATACGACTCACTATAGGGACTATTGATGGCCTTCCGACC-3' (5' primer) (SEQ ID NO: 10)
5'-CCCGGATCCTCTTTACCTCTGTGTG-3' (3' primer) (SEQ ID NO: 11)
where the small-case letters in the template oligo indicate that at each position that a mixture of reagents were used in synthesis by an amount of 62.5% of the small case letter, and 12.5% each of the other three nucleotides. Listed below the 6a sequence are the sequences of 38 isolates cloned after six rounds of SELEX performed with Rev protein with this population of RNA. The differences found in these isolates from the 6a sequences are indicated by bold-faced characters. Underlined are the predicted base pairings that comprise the bulge-flanking stems of the Motif I Rev ligands. Bases that are included from the 5' and 3' fixed flanking sequences are lower case.
FIG. 22 shows three sets of tabulations containing:
A) The count of each nucleotide found at corresponding positions of the Rev 6a ligand sequence in the collection of sequences found in FIG. 21;
B) The fractional frequency of each nucleotide found at these positions (x÷38, where x is the count from 1.); and
C) The difference between the fractional frequency of B) and the expected frequency based on the input mixture of oligonucleotides during template synthesis [for "wild type" positions, (x÷38)-0.625 and for alternative sequences (x÷38)-0.125].
FIG. 23 shows three sets of tabulations containing:
A) The count of each base pair found at corresponding positions of the Rev 6a ligand sequence in the collection of sequences found in FIG. 21,
B) The fractional frequency of each nucleotide found at these positions (x÷38, where x is the count from A),
C) The difference between the fractional frequency of B) and the expected frequency based on the input mixture of oligonucleotides during template synthesis [for "wild type" positions, (x÷38)-0.39; for base pairs that contain one alternate nucleotide and one wild type nucleotide, (x÷38)-0.078; and for base pairings of two alternate nucleotides (x÷38)-0.016]. Values are shown for purine pyrimidine pairings only, the other eight pyrimidine and purine pairings are collectively counted and shown as "other" and are computed for section
C) as (x÷38)-0.252.
FIG. 24A shows the previously determined Rev protein ligand Motif I consensus from U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991.
FIG. 24B shows the 6a sequence from the same application.
FIG. 24C shows the preferred consensus derived from the biased randomization SELEX as interpreted from the data presented in FIGS. 22 and 23. Absolutely conserved positions in the preferred consensus are shown in bold face characters, and S-S' indicates either a C-G or G-C base pair.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
This application is an extension and improvement of the method for identifying nucleic acid ligands referred to as SELEX. The SELEX method is described in detail in U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991 entitled Nucleic Acid Ligands and 07/536,428 filed June 11, 1990 entitled Systematic Evolution of Ligands by Exponential Enrichment now abandoned. The full text of these applications, including but not limited to, all definitions and descriptions of the SELEX process, are specifically incorporated herein by reference.
This application includes methods for identifying and producing improved nucleic acid ligands based on the basic SELEX process. The application includes separate sections covering the following embodiments of the invention: I. The SELEX Process; II. Techniques for Identifying Improved Nucleic Acid Ligands Subsequent to Performing SELEX; III. Sequential SELEX Experiments--Walking; IV. Elucidation of Structure of Ligands Via Covariance Analysis; V. Elucidation of an Improved Nucleic Acid Ligand for HIV-RT; VI. Performance of Walking Experiment With HIV-RT Nucleic Acid Ligand to Identify Extended Nucleic Acid Ligands; and VII. Elucidation of an Improved Nucleic Acid Ligand for HIV-1 Rev Protein.
Improved nucleic acid ligands to the HIV-RT and HIV-1 Rev proteins are disclosed and claimed herein. This invention includes the specific nucleic acid ligands identified herein. The scope of the ligands covered by the invention extends to all ligands of the HIV-RT and Rev proteins identified according to the procedures described herein. More specifically, this invention includes nucleic acid sequences that are substantially homologous to and that have substantially the same ability to bind the HIV-RT or Rev proteins, under physiological conditions, as the nucleic acid ligands identified herein. By substantially homologous, it is meant, a degree of homology in excess of 70%, most preferably in excess of 80%. Substantially homologous also includes base pair flips in those areas of the nucleic acid ligands that include base pairing regions. Substantially the same ability to bind the HIV-RT or Rev protein means that the affinity is within two orders of magnitude of the affinity of the nucleic acid ligands described herein. It is well within the skill of those of ordinary skill in the art to determine whether a given sequence is substantially homologous to and has substantially the same ability to bind the HIV-RT or HIV-1 Rev protein as the sequences identified herein.
I. The SELEX Process
In its most basic form, the SELEX process may be defined by the following series of steps:
1) A candidate mixture of nucleic acids of differing sequence is prepared. The candidate mixture generally includes regions of fixed sequences (i.e., each of the members of the candidate mixture contains the same sequences in the same location) and regions of randomized sequences. The fixed sequence regions are selected either: a) to assist in the amplification steps described below; b) to facilitate mimicry of a sequence known to bind to the target; or c) to enhance the concentration of a given structural arrangement of the nucleic acids in the candidate mixture. The randomized sequences can be totally randomized (i.e., the probability of finding a base at any position being one in four) or only partially randomized (e.g., the probability of finding a base at any location can be selected at any level between 0 and 100 percent).
2) The candidate mixture is contacted with the selected target under conditions favorable for binding between the target and members of the candidate mixture. Under these circumstances, the interaction between the target and the nucleic acids of the candidate mixture can be considered as forming nucleic acid-target pairs between the target and the nucleic acids having the strongest affinity for the target.
3) The nucleic acids with the highest affinity for the target are partitioned from those nucleic acids with lesser affinity to the target. Because only an extremely small number of sequences (and possibly only one molecule of nucleic acid) corresponding to the highest affinity nucleic acids exist in the candidate mixture, it is generally desirable to set the partitioning criteria so that a significant amount of the nucleic acids in the candidate mixture (approximately 5-50%) are retained during partitioning.
4) Those nucleic acids selected during partitioning as having the relatively higher affinity to the target are then amplified to create a new candidate mixture that is enriched in nucleic acids having a relatively higher affinity for the target.
5) By repeating the partitioning and amplifying steps above, the newly formed candidate mixture contains fewer and fewer unique sequences, and the average degree of affinity of the nucleic acids to the target will generally increase. Taken to its extreme, the SELEX process will yield a candidate mixture containing one or a small number of unique nucleic acids representing those nucleic acids from the original candidate mixture having the highest affinity to the target molecule.
The SELEX Patent Applications describe and elaborate on this process in great detail. Included are targets that can be used in the process; methods for the preparation of the initial candidate mixture; methods for partitioning nucleic acids within a candidate mixture; and methods for amplifying partitioned nucleic acids to generate enriched candidate mixtures. The SELEX Patent Applications also describe ligand solutions obtained to a number of target species, including both protein targets wherein the protein is and is not a nucleic acid binding protein.
SELEX delivers high affinity ligands of a target molecule. This represents a singular achievement that is unprecedented in the field of nucleic acids research. The present invention is directed at methods for taking the SELEX derived ligand solution in order to develop novel nucleic acid ligands having the desired characteristics. The desired characteristics for a given nucleic acid ligand may vary. All nucleic acid ligands are capable of forming a complex with the target species. In some cases, it is desired that the nucleic acid ligand will serve to inhibit one or more of the biological activities of the target. In other cases, it is desired that the nucleic acid ligand serves to modify one or more of the biological activities of the target. In other cases, the nucleic acid ligand serves to identify the presence of the target, and its effect on the biological activity of the target is irrelevant.
II. Techniques for Identifying Improved Nucleic Acid Ligands Subsequent to Performing SELEX
In order to produce nucleic acids desirable for use as a pharmaceutical, it is preferred that the nucleic acid ligand 1) binds to the target in a manner capable of achieving the desired effect on the target; 2) be as small as possible to obtain the desired effect; 3) be as stable as possible; and 4) be a specific ligand to the chosen target. In most, if not all, situations it is preferred that the nucleic acid ligand have the highest possible affinity to the target. Modifications or derivatizations of the ligand that confer resistance to degradation and clearance in situ during therapy, the capability to cross various tissue or cell membrane barriers, or any other accessory properties that do not significantly interfere with affinity for the target molecule may also be provided as improvements. The present invention includes the methods for obtaining improved nucleic acid ligands after SELEX has been performed.
Assays of ligand effects on target molecule function. One of the uses of nucleic acid ligands derived by SELEX is to find ligands that alter target molecule function. Because ligand analysis requires much more work than is encountered during SELEX enrichments, it is a good procedure to first assay for inhibition or enhancement of function of the target protein. One could even perform such functional tests of the combined ligand pool prior to cloning and sequencing. Assays for the biological function of the chosen target are generally available and known to those skilled in the art, and can be easily performed in the presence of the nucleic acid ligand to determine if inhibition occurs.
Affinity assays of the ligands. SELEX enrichment will supply a number of cloned ligands of probable variable affinity for the target molecule. Sequence comparisons may yield consensus secondary structures and primary sequences that allow grouping of the ligand sequences into motifs. Although a single ligand sequence (with some mutations) can be found frequently in the total population of cloned sequences, the degree of representation of a single ligand sequence in the cloned population of ligand sequences may not absolutely correlate with affinity for the target molecule. Therefore mere abundance is not the sole criterion for judging "winners" after SELEX and binding assays for various ligand sequences (adequately defining each motif that is discovered by sequence analysis) are required to weigh the significance of the consensus arrived at by sequence comparisons. The combination of sequence comparison and affinity assays should guide the selection of candidates for more extensive ligand characterization.
Information boundaries determination. An important avenue for narrowing down what amount of sequence is relevant to specific affinity is to establish the boundaries of that information within a ligand sequence. This is conveniently accomplished by selecting end-labeled fragments from hydrolyzed pools of the ligand of interest so that 5' and 3' boundaries of the information can be discovered. To determine a 3' boundary, one performs a large-scale in vitro transcription of the PCRd ligand, gel purifies the RNA using UV shadowing on an intensifying screen, phosphatases the purified RNA, phenol extracts extensively, labels by kinasing with 32P, and gel purifies the labeled product (using a film of the gel as a guide). The resultant product may then be subjected to pilot partial digestions with RNase T1 (varying enzyme concentration and time, at 50° C. in a buffer of 7M urea, 50 mM NaCitrate pH 5.2) and alkaline hydrolysis (at 50 mM NaCO3, adjusted to pH 9.0 by prior mixing of 1M bicarbonate and carbonate solutions; test over ranges of 20 to 60 minutes at 95° C.). Once optimal conditions for alkaline hydrolysis are established (so that there is an even distribution of small to larger fragments) one can scale up to provide enough material for selection by the target (usually on nitrocellulose filters). One then sets up binding assays, varying target protein concentration from the lowest saturating protein concentration to that protein concentration at which approximately 10% of RNA is bound as determined by the binding assays for the ligand. One should vary target concentration (if target supplies allow) by increasing volume rather than decreasing absolute amount of target; this provides a good signal to noise ratio as the amount of RNA bound to the filter is limited by the absolute amount of target. The RNA is eluted as in SELEX and then run on a denaturing gel with T1 partial digests so that the positions of hydrolysis bands can be related to the ligand sequence.
The 5' boundary can be similarly determined. Large-scale in vitro transcriptions are purified as described above. There are two methods for labeling the 3' end of the RNA. One method is to kinase Cp with 32P (or purchase 32P-Cp) and ligate to the purified RNA with RNA ligase. The labeled RNA is then purified as above and subjected to very identical protocols. An alternative is to subject unlabeled RNAs to partial alkaline hydrolyses and extend an annealed, labeled primer with reverse transcriptase as the assay for band positions. One of the advantages over pCp labeling is the ease of the procedure, the more complete sequencing ladder (by dideoxy chain termination sequencing) with which one can correlate the boundary, and increased yield of assayable product. A disadvantage is that the extension on eluted RNA sometimes contains artifactual stops, so it may be important to control by spotting and eluting starting material on nitrocellulose filters without washes and assaying as the input RNA.
The result is that it is possible to find the boundaries of the sequence information required for high affinity binding to the target.
An instructive example is the determination of the boundaries of the information found in the nucleic acid ligand for HIV-RT. (See, U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991.) These experiments are described in detail below. The original pool of enriched RNAs yielded a few specific ligands for HIV-RT (one ligand, 1.1, represented 1/4 of the total population, nitrocellulose affinity sequences represented 1/2 and some RNAs had no affinity for either). Two high-affinity RT ligands shared the sequence . . . UUCCGNNNNNNNNCGGGAAAA (SEQ ID NO: 1) . . . Boundary experiments of both ligands established a clear 3' boundary and a less clear 5' boundary. It can be surmised from the boundary experiments and secondary SELEX experiments that the highest affinity ligands contained the essential information UCCGNNNNNNNNCGGGAAAAN'N'N'N' (SEQ ID NO:2) (where N's base pair to Ns in the 8 base loop sequence of the hairpin formed by the pairing of UCCG to CGGG) and that the 5' U would be dispensable with some small loss in affinity. In this application, the construction of model compounds confirmed that there was no difference in the affinity of sequences with only one 5' U compared to 2 5' U's (as is shared by the two compared ligands), that removal of both U's caused a 5-fold decrease in affinity and of the next C a more drastic loss in affinity. The 3' boundary which appeared to be clear in the boundary experiments was less precipitous. This new information can be used to deduce that what is critical at the 3' end is to have at least three base-paired nucleotides (to sequences that loop between the two strands of Stem 1). Only two base-paired nucleotides result in a 12-fold reduction in affinity. Having no 3' base-paired nucleotides (truncation at the end of Loop 2) results in an approximately 70-fold reduction in affinity.
Quantitative and Qualitative Assessment of Individual Nucleotide Contributions to Affinity
SECONDARY SELEX. Once the minimal high affinity ligand sequence is identified, it may be useful to identify the nucleotides within the boundaries that are crucial to the interaction with the target molecule. One method is to create a new random template in which all of the nucleotides of a high affinity ligand sequence are partially randomized or blocks of randomness are interspersed with blocks of complete randomness. Such "secondary" SELEXes produce a pool of ligand sequences in which crucial nucleotides or structures are absolutely conserved, less crucial features preferred, and unimportant positions unbiased. Secondary SELEXes can thus help to further elaborate a consensus that is based on relatively few ligand sequences. In addition, even higher-affinity ligands may be provided whose sequences were unexplored in the original SELEX.
In this application we show such a biased randomization for ligands of the HIV-1 Rev protein. In U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991, nucleic acid ligands to the HIV-1 Rev protein were described. One of these ligand sequences bound with higher affinity than all of the other ligand sequences (Rev ligand sequence 6a, shown in FIG. 12) but existed as only two copies in the 53 isolates that were cloned and sequenced. In this application, this sequence was incorporated in a secondary SELEX experiment in which each of the nucleotides of the 6a sequence (confined to that part of the sequence which comprises a Rev protein binding site defined by homology to others of Rev ligand motif I) was mixed during oligonucleotide synthesis with the other three nucleotides in the ratio 62.5:12.5:12.5:12.5. For example, when the sequence at position G1 is incorporated during oligo synthesis, the reagents for G,A,T, and C are mixed in the ratios 62.5:12.5:12.5:12.5. After six rounds of SELEX using the Rev protein, ligands were cloned from this mixture so that a more comprehensive consensus description could be derived.
NUCLEOTIDE SUBSTITUTION. Another method is to test oligo-transcribed variants where the SELEX consensus may be confusing. As shown above, this has helped us to understand the nature of the 5' and 3' boundaries of the information required to bind HIV-RT. As is shown in the attached example this has helped to quantitate the consensus of nucleotides within Stem 1 of the HIV-RT pseudoknot.
CHEMICAL MODIFICATION. Another useful set of techniques are inclusively described as chemical modification experiments. Such experiments may be used to probe the native structure of RNAs, by comparing modification patterns of denatured and non-denatured states. The chemical modification pattern of an RNA ligand that is subsequently bound by target molecule may be different from the native pattern, indicating potential changes in structure upon binding or protection of groups by the target molecule. In addition, RNA ligands will fail to be bound by the target molecule when modified at positions crucial to either the bound structure of the ligand or crucial to interaction with the target molecule. Such experiments in which these positions are identified are described as "chemical modification interference" experiments.
There are a variety of available reagents to conduct such experiments that are known to those skilled in the art (see, Ehresmann et al., Nuc. Acids. Res., 15:9109-9128,(1987)). Chemicals that modify bases can be used to modify ligand RNAs. A pool is bound to the target at varying concentrations and the bound RNAs recovered (much as in the boundary experiments) and the eluted RNAs analyzed for the modification. Assay can be by subsequent modification-dependent base removal and aniline scission at the baseless position or by reverse transcription assay of sensitive (modified) positions. In such assays bands (indicating modified bases) in unselected RNAs appear that disappear relative to other bands in target protein-selected RNAs. Similar chemical modifications with ethylnitrosourea, or via mixed chemical or enzymatic synthesis with, for example, 2'-methoxys on ribose or phosphorothioates can be used to identify essential atomic groups on the backbone. In experiments with 2'-methoxy vs. 2'-OH mixtures, the presence of an essential OH group results in enhanced hydrolysis relative to other positions in molecules that have been stringently selected by the target.
An example of how chemical modification can be used to yield useful information about a ligand and help efforts to improve its functional stability is given below for HIV-RT. Ethylnitrosourea modification interference identified 5 positions at which modification interfered with binding and 2 of those positions at which it interfered drastically. Modification of various atomic groups on the bases of the ligand were also identified as crucial to the interaction with HIV-RT. Those positions were primarily in the 5' helix and bridging loop sequence that was highly conserved in the SELEX phylogeny (Stem I and Loop 2, FIG. 1). These experiments not only confirmed the validity of that phylogeny, but informed ongoing attempts to make more stable RNAs. An RT ligand was synthesized in which all positions had 2'-methoxy at the ribose portions of the backbone. This molecule bound with drastically reduced affinity for HIV-RT. Based on the early modification interference experiments and the SELEX phylogeny comparisons, it could be determined that the 3' helix (Stem II FIG. 1) was essentially a structural component of the molecule. A ligand in which the 12 ribose residues of that helix were 2'-methoxy was then synthesized and it bound with high affinity to HIV-RT. In order to determine if any specific 2'-OHs of the remaining 14 residues were specifically required for binding, a molecule in which all of the riboses of the pseudoknot were synthesized with mixed equimolar (empirically determined to be optimal) reagents for 2'-OH and 2'-methoxy formation. Selection by HIV-RT from this mixture followed by alkaline hydrolysis reveals bands of enhanced hydrolysis indicative of predominating 2' hydroxyls at those positions. Analysis of this experiment lead to the conclusion that residues (G4, A5, C13 and G14) must have 2'-OH for high affinity binding to HIV-RT.
Comparisons of the intensity of bands for bound and unbound ligands may reveal not only modifications that interfere with binding, but also modifications that enhance binding. A ligand may be made with precisely that modification and tested for the enhanced affinity. Thus chemical modification experiments can be a method for exploring additional local contacts with the target molecule, just as "walking" (see below) is for additional nucleotide level contacts with adjacent domains.
One of the products of the SELEX procedure is a consensus of primary and secondary structures that enables the chemical or enzymatic synthesis of oligonucleotide ligands whose design is based on that consensus. Because the replication machinery of SELEX requires that rather limited variation at the subunit level (ribonucleotides, for example), such ligands imperfectly fill the available atomic space of a target molecule's binding surface. However, these ligands can be thought of as high-affinity scaffolds that can be derivatized to make additional contacts with the target molecule. In addition, the consensus contains atomic group descriptors that are pertinent to binding and atomic group descriptors that are coincidental to the pertinent atomic group interactions. For example, each ribonucleotide of the pseudoknot ligand of HIV-RT contains a 2' hydroxyl group on the ribose, but only two of the riboses of the pseudoknot ligand cannot be substituted at this position with 2'-methoxy. A similar experiment with deoxyribonucleotide mixtures with ribonucleotide mixtures (as we have done with 2'-methoxy and 2' hydroxy mixtures) would reveal which riboses or how many riboses are dispensable for binding HIV-RT. A similar experiment with more radical substitutions at the 2' position would again reveal the allowable substitutions at 2' positions. One may expect by this method to find derivatives of the pseudoknot ligand that confer higher affinity association with HIV-RT. Such derivatization does not exclude incorporation of cross-linking agents that will give specifically directly covalent linkages to the target protein. Such derivatization analyses are not limited to the 2' position of the ribose, but could include derivatization at any position in the base or backbone of the nucleotide ligand.
A logical extension of this analysis is a situation in which one or a few nucleotides of the polymeric ligand is used as a site for chemical derivative exploration. The rest of the ligand serves to anchor in place this monomer (or monomers) on which a variety of derivatives are tested for non-interference with binding and for enhanced affinity. Such explorations may result in small molecules that mimic the structure of the initial ligand framework, and have significant and specific affinity for the target molecule independent of that nucleic acid framework. Such derivatized subunits, which may have advantages with respect to mass production, therapeutic routes of administration, delivery, clearance or degradation than the initial SELEX ligand, may become the therapeutic and may retain very little of the original ligand. This approach is thus an additional utility of SELEX. SELEX ligands can allow directed chemical exploration of a defined site on the target molecule known to be important for the target function.
Structure determination. These efforts have helped to confirm and evaluate the sequence and structure dependent association of ligands to HIV-RT. Additional techniques may be performed to provide atomic level resolution of ligand/target molecule complexes. These are NMR spectroscopy and X-ray crystallography. With such structures in hand, one can then perform rational design as improvements on the evolved ligands supplied by SELEX. The computer modeling of nucleic acid structures is described below.
Chemical Modification. This invention includes nucleic acid ligands wherein certain chemical modifications have been made in order to increase the in vivo stability of the ligand or to enhance or mediate the delivery of the ligand. Examples of such modifications include chemical substitutions at the ribose and/or phosphate positions of a given RNA sequence. See, e.g., Cook, et al. PCT Application WO 9203568; U.S. Pat. No. 5,118,672 of Schinazi et al.; Hobbs et al. Biochem 12:5138 (1973); Guschlbauer et al. Nucleic Acids Res. 4:1933 (1977); Shibahara et al. Nucl. Acids. Res. 15:4403 (1987); Pieken et al. Science 253:314 (1991), each of which is specifically incorporated herein by reference.
III. Sequential SELEX Experiments--Walking
In one embodiment of this invention, after a minimal consensus ligand sequence has been determined for a given target, it is possible to add random sequence to the minimal consensus ligand sequence and evolve additional contacts with the target, perhaps to separate but adjacent domains. This procedure is referred to as "walking" in the SELEX Patent Applications. The successful application of the walking protocol is presented below to develop an enhanced binding ligand to HIV-RT.
The walking experiment involves two SELEX experiments performed sequentially. A new candidate mixture is produced in which each of the members of the candidate mixture has a fixed nucleic acid region that corresponds to a SELEX-derived nucleic acid ligand. Each member of the candidate mixture also contains a randomized region of sequences. According to this method it is possible to identify what are referred to as "extended" nucleic acid ligands, that contain regions that may bind to more than one binding domain of a target.
IV. Elucidation of Structure of Ligands Via Covariance Analysis
In conjunction with the empirical methods for determining the three dimensional structure of nucleic acids, the present invention includes computer modeling methods for determining structure of nucleic acid ligands.
Secondary structure prediction is a useful guide to correct sequence alignment. It is also a highly useful stepping-stone to correct 3D structure prediction, by constraining a number of bases into A-form helical geometry.
Tables of energy parameters for calculating the stability of secondary structures exist. Although early secondary structure prediction programs attempted to simply maximize the number of base-pairs formed by a sequence, most current programs seek to find structures with minimal free energy as calculated by these thermodynamic parameters. There are two problems in this approach. First, the thermodynamic rules are inherently inaccurate, typically to 10% or so, and there are many different possible structures lying within 10% of the global energy minimum. Second, the actual secondary structure need not lie at a global energy minimum, depending on the kinetics of folding and synthesis of the sequence. Nonetheless, for short sequences, these caveats are of minor importance because there are so few possible structures that can form.
The brute force predictive methods is a dotplot: make an N by N plot of the sequence against itself, and mark an X everywhere a basepair is possible. Diagonal runs of X's mark the location of possible helices. Exhaustive tree-searching methods can then search for all possible arrangements of compatible (i.e., non-overlapping) helices of length L or more; energy calculations may be done for these structures to rank them as more or less likely. The advantages of this method are that all possible topologies, including pseudoknotted conformations, may be examined, and that a number of suboptimal structures are automatically generated as well. The disadvantages of the method are that it can run in the worst cases in time proportional to an exponential factor of the sequence size, and may not (depending on the size of the sequence and the actual tree search method employed) look deep enough to find a global minimum.
The elegant predictive method, and currently the most used, is the Zuker program. Zuker (1989) Science 244:48-52. Originally based on an algorithm developed by Ruth Nussinov, the Zuker program makes a major simplifying assumption that no pseudoknotted conformations will be allowed. This permits the use of a dynamic programming approach which runs in time proportional to only N3 to N4, where N is the length of the sequence. The Zuker program is the only program capable of rigorously dealing with sequences of than a few hundred nucleotides, so it has come to be the most commonly used by biologists. However, the inability of the Zuker program to predict pseudoknotted conformations is a fatal flaw, in that several different SELEX experiments so far have yielded pseudoknotted RNA structures, which were recognized by eye. A brute-force method capable of predicting pseudoknotted conformations must be used.
The central element of the comparative sequence analysis of the present invention is sequence covariations. A covariation is when the identity of one position depends on the identity of another position; for instance, a required Watson-Crick base pair shows strong covariation in that knowledge of one of the two positions gives absolute knowledge of the identity at the other position. Covariation analysis has been used previously to predict the secondary structure of RNAs for which a number of related sequences sharing a common structure exist, such as tRNA, rRNAs, and group I introns. It is now apparent that covariation analysis can be used to detect tertiary contacts as well.
Stormo and Gutell (Gutell et al., Nuc. Acids Res. 20:5785-5795 (1992) have designed and implemented an algorithm that precisely measures the amount of covariations between two positions in an aligned sequence set. The program is called "MIXY"--Mutual Information at position X and Y.
Consider an aligned sequence set. In each column or position, the frequency of occurrence of A, C, G, U, and gaps is calculated. Call this frequency f(bx), the frequency of base b in column x. Now consider two columns at once. The frequency that a given base b appears in column x is f(bx) and the frequency that a given base b appears in column y is f(by). If position x and position y do not care about each other's identity--that is, the positions are independent; there is no covariation--the frequency of observing bases bx and by at position x and y in any given sequence should be just f(bx by)=f(bx)f(by). If there are substantial deviations of the observed frequencies of pairs from their expected frequencies, the positions are said to covary. The amount of deviation from expectation may be quantified with an information measure M(x,y), the mutual information of x and y: ##EQU1## M(x,y) can be described as the number of bits of information one learns about the identity of position y from knowing just the identity of position y from knowing just the identity of position x. If there is no covariation, M(x,y) is zero; larger values of M(x,y) indicate strong covariation.
These numbers correlated extremely well to a probability for close physical contact in the tertiary structure, when this procedure was applied to the tRNA sequence data set. The secondary structure is extremely obvious as peaks in the M(x,y) values, and most of the tertiary contacts known from the crystal structure appear as peaks as well.
These covariation values may be used to develop three-dimensional structural predictions.
In some ways, the problem is similar to that of structure determination by NMR. Unlike crystallography, which in the end yields an actual electron density map, NMR yields a set of interatomic distances. Depending on the number of interatomic distances one can get, there may be one, few, or many 3D structures with which they are consistent. Mathematical techniques had to be developed to transform a matrix of interatomic distances into a structure in 3D space. The two main techniques in use are distance geometry and restrained molecular dynamics.
Distance geometry is the more formal and purely mathematical technique. The interatomic distances are considered to be coordinates in an N-dimensional space, where N is the number of atoms. In other words, the "position" of an atom is specified by N distances to all the other atoms, instead of the three (x,y,z) that we are used to thinking about. Interatomic distances between every atom are recorded in an N by N distance matrix. A complete and precise distance matrix is easily transformed into a 3 by N Cartesian coordinates, using matrix algebra operations. The trick of distance geometry as applied to NMR is dealing with incomplete (only some of the interatomic distances are known) and imprecise data (distances are known to a precision of only a few angstroms at best). Much of the time of distance geometry-based structure calculation is thus spent in pre-processing the distance matrix, calculating bounds for the unknown distance values based on the known ones, and narrowing the bounds on the known ones. Usually, multiple structures are extracted from the distance matrix which are consistent with a set of NMR data; if they all overlap nicely, the data were sufficient to determine a unique structure. Unlike NMR structure determination, covariance gives only imprecise distance values, but also only probabilistic rather than absolute knowledge about whether a given distance constraint should be applied.
Restrained molecular dynamics is a more ad hoc procedure. Given an empirical force field that attempts to describe the forces that all the atoms feel (van der Waals, covalent bonding lengths and angles, electrostatics), one can simulate a number of femtosecond time steps of a molecule's motion, by assigning every atom at a random velocity (from the Boltzmann distribution at a given temperature) and calculating each atom's motion for a femtosecond using Newtonian dynamical equations; that is "molecular dynamics". In restrained molecular dynamics, one assigns extra ad hoc forces to the atoms when they violate specified distance bounds.
In the present case, it is fairly easy to deal with the probabilistic nature of data with restrained molecular dynamics. The covariation values may be transformed into artificial restraining forces between certain atoms for certain distance bounds; varying the magnitude of the force according to the magnitude of the covariance.
NMR and covariance analysis generates distance restraints between atoms or positions, which are readily transformed into structures through distance geometry or restrained molecular dynamics. Another source of experimental data which may be utilized to determine the three dimensional structures of nucleic acids is chemical and enzymatic protection experiments, which generate solvent accessibility restraints for individual atoms or positions.
V. ELUCIDATION OF AN IMPROVED NUCLEIC ACID LIGAND FOR HIV-RT
An example of the methods of the present invention are presented herein for the nucleic acid ligand for HIV-1 reverse transcriptase (HIV-RT). U.S. patent application Ser. No. 07/714,131 describes the results obtained when SELEX was performed with the HIV-RT target. Inspection of the nucleic acid sequences that were found to have a high affinity to HIV-RT, it was concluded that the nucleic acid ligand solution was configured as a pseudoknot.
Described herein are experiments which establish the minimum number of sequences necessary to represent the nucleic acid ligand solution via boundary studies. Also described are the construction of variants of the ligand solution which are used to evaluate the contributions of individual nucleotides in the solution to the binding of the ligand solution to HIV-RT. Also described is the chemical modification of the ligand solution; 1) to corroborate its predicted pseudoknot structure; 2) to determine which modifiable groups are protected from chemical attack when bound to HIV-RT (or become unprotected during binding); and 3) to determine what modifications interfere with binding to HIV-RT (presumably by modification of the three dimensional structure of the ligand solution) and, therefore, which are presumably involved in the proximal contacts with the target.
The nucleic acid ligand solution previously determined is shown in FIG. 1. Depicted is an RNA pseudoknot in which Stem 1 (as labeled) is conserved and Stem 2 is relatively non-conserved; X indicates no conservation and X' base-pairs to X. In the original SELEX consensus U1 was preferred (existing at this relative position in 11 of the 18 sequences that contributed to the consensus), but A1 was also found frequently (in 6 of the 18). There were two sequences in which C-G was substituted for the base-pair of G4-C13 and one A-U substitution. The preferred number of nucleotides connecting the two strands of Stem 1 was eight (in 8 of 18). The number and pattern of base-paired nucleotides comprising Stem 2 and the preference for A5 and A12 were derived from the consensus of a secondary SELEX in which the random region was constructed as follows NNUUCCGNNNNNNNNCGGGAAAANNNN (SEQ ID NO:3) (Ns are randomized). One of the ligands was found to significantly inhibit HIV-RT and failed to inhibit AMV or MMLV reverse transcriptases.
Refinement of the information boundaries. The first two SELEX experiments in which 32 nucleotide positions were randomized provided high affinity ligands in which there was variable length for Stem 1 at its 5' end; that is, some ligands had the sequence UUCCG which could base pair to CGGGA, UCCG to CGGG or CCG to CGG. Determination of the boundaries of the sequences donating high-affinity to the interaction with HIV-RT was accomplished by selection from partial alkaline hydrolysates of end-labeled clonal RNAs, a rapid but qualitative analysis which suggested that the highest affinity ligands contained the essential information UCCGNNNNNNNNCGGGAAAAN'N'N'N' (SEQ ID NO: 2) (where N's base pair to Ns in the 8 base loop sequence of the hairpin formed by the pairing of UCCG to CGGG) and that the 5' U would be dispensable with some small loss in affinity. In order to more stringently test the 5' sequences in a homogeneous context, the binding experiments depicted in FIG. 2 were performed. The RNA's transcribed from oligonucleotide templates were all the same as the complete sequence shown in the upper right hand corner of the figure, except for the varying 5' ends as shown in the boxes A-E lining the left margin. The result is that one 5' U is sufficient for the highest-affinity binding to HIV-RT (boxes A and B), that with no U there is reduced binding (box C), and that any further removal of 5' sequences reduces binding to that of non-specific sequences (box D). The design (hereafter referred to as ligand B) with only one 5' U (U 1) was used for the rest of the experiments described here.
Dependence on the length of Stem 2 was also examined by making various 3' truncations at the 3' end of ligand B. Deletion of as many as 3 nucleotides from the 3' end (A24-U26) made no difference in affinity of the molecule for HIV-RT. Deletion of the 3'-terminal 4 nucleotides (C23-U26) resulted in 7-fold reduced binding, of 5 (G22-U26) resulted in approximately 12-fold reduction and of 6 nucleotides (U21-U26, or no 3' helix) an approximately 70-fold reduction in affinity. Such reductions were less drastic than reductions found for single-base substitutions reported below, suggesting (with other data reported below) that this helix serves primarily a structural role that aids the positioning of crucial groups in Loop 2.
Testing the SELEX consensus for Stem 1. Various nucleotide substitutions in the conserved Stem 1 were prepared and their affinity to HIV-RT determined. As shown in FIG. 3, substitution of an A for U1 in model RNAs made little difference in affinity for HIV-RT. C (which would increase the stability of Steml) or G (represented by the U deletion experiment above) at this position resulted in approximately 20-fold lowering in affinity. Substitution of A for G16 (which would base-pair to U1) abolished specific binding. A G-C pair was substituted for C2-G15 which also abolished binding and for C3-G14 which reduced binding about 10-fold. These two positions were highly conserved in the phylogeny of SELEX ligands. Various combinations were substituted for the G4-C13 base pair. The order of affect of these on affinity were G4-C13=C-G>U-A>A-U>>>>A-C where A-U is about 20-fold reduced in affinity compared to G4-C13 and A-C is at least 100- fold reduced. These results are consistent with the SELEX consensus determined previously.
Chemical probing of the pseudoknot structure. A number of chemical modification experiments were conducted to probe the native structure of ligand B, to identify chemical modifications that significantly reduced affinity of ligand B for HIV-RT, and to discover changes in structure that may accompany binding by HIV-RT. The chemicals used were ethylnitrosourea (ENU) which modifies phosphates, dimethyl sulfate (DMS) which modifies the base-pairing faces of C (at N3) and A (at N1), carbodiimide (CMCT) which modifies the base-pairing face of U (at N3) and to some extent G (at N1), diethylpyrocarbonate (DEPC) which modifies N7 of A and to a lesser extent the N7 of G, and kethoxal which modifies the base-pairing N1 and N2 of G. Most of the assays of chemical modification were done on a ligand B sequence which was lengthened to include sequences to which a labeled primer could be annealed and extended with AMV reverse transcriptase. Assay of ENU or DEPC modified positions were done on ligand B by respective modification-dependent hydrolysis, or modified base removal followed by aniline scission of the backbone at these sites.
The results of probing the native structure as compared to modification of denatured ligand B are summarized in FIG. 4. The pattern of ENU modification was not different between denatured native states of the ligand suggesting that there is no stable involvement of the phosphates or N7 positions of purines in the solution structure of the pseudoknot. The other modification data suggest that Stem 2 forms rather stably and is resistant to any chemical modifications affecting the base-pairs shown, although the terminal A6-U26 is somewhat sensitive to modification indicating equilibration between base-paired and denatured states at this position. The single-stranded As (A5,A17,A18, A19,and A20) are fully reactive with DMS although A5, A19, and A20 are diminished in reactivity to DEPC. The base-pairs of Stem 1 seem to exhibit a gradation of resistance to modification such that G4-C13>C3-G14>C2-G15>U1-G16 where G4-C13 is completely resistant to chemical modification and U1-G16 is highly reactive. This suggests that this small helix of the pseudoknot undergoes transient and directional denaturation or "fraying".
Protection of ligand B from chemical modification bY HIV-RT. Binding of protein changes the fraying character of Helix I as shown in FIG. 5 either by stabilizing or protecting it. The natively reactive U1 is also protected upon binding. Binding of protein increases the sensitivity of the base-pair A6-U26 suggesting that this is unpaired in the bound state. This may be an indication of insufficient length of a single nucleotide Loop I during binding, either because it cannot bridge the bound Stem 1 to the end of Stem 2 in the native pseudoknot recognized by RT or because binding increases the length requirement of Loop I by changing the conformation from the native state. A17 and A19 of Loop II are also protected by binding to HIV-RT. In addition, the single base bridge A12 is protected upon binding.
Modification interference studies of the RT ligand B. The RNA ligand B was partially modified (with all of the chemicals mentioned above for structure determination). This modified population was bound with varying concentrations of the protein, and the bound species were assayed for the modified positions. From this, it can be determined where modification interferes with binding, and where there is no or little effect. A schematic diagram summarizing these modification interference results is shown in FIG. 6. As shown, most of the significant interference with binding is clustered on the left hand side of the pseudoknot which contains the Stem 1 and Loop 2. This is also the part of the molecule that was highly conserved (primary sequence) in the collection of sequences isolated by SELEX and where substitution experiments produced the most drastic reduction in binding affinity to HIV-RT.
Substitution of 2'-methoxy for 2'-hydroxyl on riboses of ligand B. "RNA" molecules in which there is a 2'-methoxy bonded to the 2' carbon of the ribose instead of the normal hydroxyl group are resistant to enzymatic and chemical degradation. In order to test how extensively 2'-methoxys can be substituted for 2'-OH's in RT ligands, four oligos were prepared as shown in FIG. 7. Because fully substituted 2'-methoxy ligand binds poorly (ligand D), and because we had found that most of the modification interference sites were clustered at one end of the pseudoknot, subsequent attempts to substitute were confined to the non-specific 3' helix as shown in boxes B and C. Both of these ligands bind with high affinity to HIV-RT. Oligonucleotides were then prepared in which the allowed substitutions at the ribose of Stem 2 were all 2'-methoxy as in C of FIG. 7 and at the remaining 14 positions mixed synthesis were done with 2'-methoxy and 2'-OH phosphoramidite reagents. These oligos were subjected to selection by HIV-RT followed by alkaline hydrolysis of selected RNAs and gel separation (2'-methoxys do not participate in alkaline hydrolysis as do 2'-hydroxyls). As judged by visual inspection of films (see FIG. 8) and quantitative determination of relative intensities using an Ambis detection system (see Example below for method of comparison), the ligands selected by HIV-RT from the mixed incorporation populations showed significantly increased hydrolysis at positions C13 and G14 indicating interference by 2'-methoxys at these positions. In a related experiment where mixtures at all positions were analyzed in this way, G4, A5, C13 and G14 showed 2' O-methyl interference.
The results of substitution experiments, quantitative boundary experiments and chemical probing experiments are highly informative about the nature of the pseudoknot inhibitor of HIV-RT and highlight crucial regions of contact on this RNA. These results are provided on a nucleotide by nucleotide basis below.
U1 can be replaced with A with little loss in affinity but not by C or G. Although U1 probably makes transient base-pairing to G16, modification of U1-N3 with CMCT does not interfere with binding to HIV-RT. However, binding by HIV-RT protects the N3 of U1 perhaps by steric or electrostatic shielding of this position. Substitution with C which forms a more stable base-pair with G16 reduces affinity. Replacement of G16 with A which forms a stable U1-A16 pair abolishes specific affinity for HIV-RT and modification of G16-N1 strongly interferes with binding to HIV-RT. This modification of G16-N1 must prevent a crucial contact with the protein. Why G substitutions for U1 reduce affinity and A substitutions do not is not clear. Admittedly the G substitution is in a context in which the 5' end of the RNA is one nucleotide shorter, however synthetic RNAs in which U1 is the 5' terminal nucleotide bind with unchanged affinity from those in vitro transcripts with two extra Gs at the 5' end (FIG. 7). Perhaps A at U1 replaces a potential U interaction with a similar or different interaction with HIV-RT a replacement that cannot be performed by C or G at this position.
The next base-pair of Stem 1 (C2-G15) cannot be replaced by a G-C base-pair without complete loss of specific affinity for HIV-RT. Modification of the base-pairing faces of either nucleotide strongly interferes with binding to HIV-RT and binding with HIV-RT protects from these modifications. Substitution of the next base-pair, C3-G14, with a G-C pair shows less drastic reduction of affinity, but modification is strongly interfering at this position. Substitution of a C-G pair for G4-C13 has no effect on binding, and substitution of the less stable A-U and U-A pairs allow some specific affinity. Substitution of the non-pairing A-C for these positions abolishes specific binding. This correlates with the appearance of C-G substitutions and one A-U substitution in the original SELEX phylogeny at this position, the non-reactivity of this base-pair in the native state, and the high degree of modification interference found for these bases.
The chemical modification data of Loop 2 corroborate well the phylogenetic conservation seen in the original SELEX experiments. Strong modification interference is seen at positions A17 and A19. Weak modification interference occurs at A20 which correlates with the finding of some Loop 2's of the original SELEX that are deleted at this relative position (although the chemical interference experiments conducted do not exhaustively test all potential contacts that a base may make with HIV-RT). A18 is unconserved in the original SELEX and modification at this position does not interfere, nor is this position protected from modification by binding to HIV-RT.
Taken together the above data suggest that the essential components of Stem 1 are a single-stranded 5' nucleotide (U or A) which may make sequence specific contact with the protein and a three base-pair helix (C2-G15, C3-G14, G4-C13) where there are sequence-specific interactions with the HIV-RT at the first two base-pairs and a preference for a strong base-pair (i.e. either C-G or G-C) at the third loop closing position of G4-C13. Loop 2 should be more broadly described as GAXAA (16-20) due to the single-stranded character of G16 which probably interacts with HIV-RT in a sequence-specific manner, as likely do A17 and A19. Stem 2 varies considerably in the pattern and number of base-pairing nucleotides, but from 3' deletion experiments reported here one could hypothesize that a minimum of 3 base-pairs in Stem 2 are required for maximal affinity. Within the context of eight nucleotides connecting the two strands comprising the helix of Stem 1, at least 2 nucleotides are required in Loop 1 of the bound ligand.
The revised ligand description for HIV-RT obtained based on the methods of this invention is shown in FIG. 11. The major differences between that shown in FIG. 1 (which is based on the original and secondary SELEX consensuses) is the length of Stem 2, the more degenerate specification of the base-pair G4-C13, the size of Loop 1 (which is directly related to the size of Stem 2) and the single-stranded character of U1 and G16.
How can these differences be reconciled? Although not limited by theory, the SELEX strategy requires 5' and 3' fixed sequences for replication. In any RNA sequence, such additional sequences increase the potential for other conformations that compete with that of the high-affinity ligand. As a result, additional structural elements that do not directly contribute to affinity, such as a lengthened Stem 2, may be selected. Given that the first two base pairs of Stem 1 must be C-G because of sequence-specific contacts the most stable closing base-pair would be G4-C13 (Freier et al., Proc. Natl. Acad. Sci. USA, 83:9373-9377 (1986)) again selected to avoid conformational ambiguity. The sequence-specific selection of U1 and G16 may be coincidental to their ability to base-pair; in other nucleic acid ligand-protein complexes such as Klenow fragment/primer-template junction and tRNA/tRNA synthetase there is significant local denaturation of base-paired nucleotides (Freemont et al., Proc. Natl. Acad. Sci. USA, 85:8924 (1988); Ronald et al., Science, 246:1135 (1989)) which may also occur in this case.
VI. Performance of Walking Experiment with HIV-RT Nucleic Acid Ligand to Identify Extended Nucleic Acid Ligands
It had previously been found that fixed sequences (of 28 nucleotides) placed 5' to the pseudoknot consensus ligand reduced the affinity to HIV-RT and that sequences (of 31 nucleotides) added 3' to the ligand increased that affinity. A SELEX experiment was therefore performed in which a 30 nucleotide variable region was added 3' to the ligand B sequence to see if a consensus of higher affinity ligands against HIV-RT could be obtained. Individual isolates were cloned and sequenced after the sixteenth round. The sequences are listed in FIG. 9 grouped in two motifs. A schematic diagram of the secondary structure and primary sequence conservation of each motif is shown in FIG. 10. The distance between the RNase H and polymerase catalytic domains of HIV-RT has recently been determined to be on the order of 18 base-pairs of an A-form RNA-DNA hybrid docked (by computer) in the pocket of a 3.5 Å resolution structure derived from X-ray crystallography (Kohlstaedt et al. Science, 256:1783-1790, (1992)). The distance from the cluster of bases determined to be crucial to this interaction in the pseudoknot and the conserved bases in the extended ligand sequence is approximately 18 base-pairs as well. Accordingly, it is concluded that the pseudoknot interacts with the polymerase catalytic site--in that the ligand has been shown to bind HIV-RT deleted for the RNAse H domain--and that the evolved extension to the pseudoknot may interact with the RNAse H domain. In general the ligands tested from each of these motifs increase affinity of the ligand B sequence to HIV-RT by at least 10-fold.
VII. ELUCIDATION OF AN IMPROVED NUCLEIC ACID LIGAND FOR HIV-1 REV PROTEIN
An example of the methods of the present invention are presented herein for the nucleic acid ligand for HIV-1 Rev protein. U.S. patent application Ser. No. 07/714,131 describes the results obtained when SELEX was performed with the Rev target. Inspection of the nucleic acid sequences that were found to have a high affinity to Rev revealed a grouping of these sequences into three Motifs (I,II, and III). Ligands of Motif I seemed to be a composite of the individual motifs described by Motifs II and III, and in general bound with higher affinity to Rev. One of the Motif I ligand sequences (Rev ligand sequence 6a) bound with significantly higher affinity than all of the ligands that were cloned and sequenced. As shown in FIG. 12, the 6a sequence is hypothesized to form a bulge between two helices with some base-pairing across this bulge.
Described herein are chemical modification experiments performed on ligand 6a designed to confirm the proposed secondary structure, find where binding of the Rev protein protects the ligand from chemical attack, and detect the nucleotides essential for Rev interaction. In addition, a secondary SELEX experiment was conducted with biased randomization of the 6a ligand sequence so as to more comprehensively describe a consensus for the highest affinity binding to the HIV-1 Rev protein.
Chemical Modification of the Rev Ligand
Chemical modification studies of the Rev ligand 6a were undertaken to determine its possible secondary structural elements, to find which modifications interfere with the binding of the ligand by Rev, to identify which positions are protected from modification upon protein binding, and to detect possible changes in ligand structure that occur upon binding.
The modifying chemicals include ethylnitrosourea (ENU) which modifies phosphates, dimethyl sulfate (DMS) which modifies the base-pairing positions N3 of C and N1 of adenine, kethoxal which modifies base-pairing positions N1 and N2 of guanine, carbodiimide (CMCT) which modifies base-paring position N3 of uracil and to a smaller extent the N1 position of guanine, and diethylpyrocarbonate (DEPC) which modifies the N7 position of adenine and to some extent also the N7 of guanine. ENU modification was assayed by modification-dependent hydrolysis of a labeled RNA chain, while all other modifying agents were used on an extended RNA ligand, with modified positions revealed by primer extension of an annealed oligonucleotide.
The chemical probing of the Rev ligand native structure is summarized in FIG. 13. The computer predicted secondary structure Zuker (1989), Science 244:48-52; Jaeger et al. (1989), Proc. Natl. Acad. Sci. USA 86:7706-7710 and native modification data are in general agreement; the ligand is composed of three helical regions, one four-base hairpin loop, and three "bulge" regions (see FIG. 13 for a definition of these structural "elements").
ENU modification of phosphates was unchanged for ligands under native and denaturing conditions, indicating no involvement of phosphate groups in the secondary or tertiary structure of the RNA. In general, all computer-predicted base-pairing regions are protected from modification. One exception is the slight modifications of N7 (G10, A11, G12) in the central helix (normally a protected position in helices). These modifications are possibly a result of helical breathing; the absence of base-pairing face modifications in the central helix suggest that the N7 accessibility is due to small helical distortions rather than a complete, local unfolding of the RNA. The G19 -U22 hairpin loop is fully modified, except for somewhat partial modification of G19.
The most interesting regions in the native structure are the three "bulge" regions, U8 -U9, A13 -A14 -A15, and G26 -A27. U8 -U9 are fully modified by CMCT, possibly indicating base orientations into solvent A13, A14 and A15 are all modified by DMS and DEPC with the strongest modifications occurring on the central A14. The bulge opposite to the A13 -A15 region shows complete protection of G26 and very slight modification of A27 by DMS. One other investigation of Rev-binding RNAs (Bartel et al. (1991) Cell 67:529-536) has argued for the existence of A:A and A:G non canonical base pairing, corresponding in the present ligand to A13 :A27 and A15 :G26. These possibilities are not ruled out by this modification data, although the isosteric A:A base pair suggested by Bartel et al. would use the N1A positions for base-pairing and would thus be resistant to DMS treatment. Also, an A:G pair would likely use either a N1A or N7A for pairing, leaving the A resistant to DMS or DEPC.
Modification interference of Rev binding. The results of the modification interference studies is summarized in FIG. 14 (quantitative data on individual modifying agents is presented in FIGS. 15 through 19). In general, phosphate and base modification binding interference is clustered into two regions of the RNA ligand. To a first approximation, these regions correspond to two separate motifs present in the SELEX experiments that preceded this present study. Phosphate modification interference is probably the most suggestive of actual sites for ligand-protein contacts, and constitutes an additional criterion for the grouping of the modification interference data into regions.
The first region is centered on U24 -G25 -G26, and includes interference due to phosphate, base-pairing face, and N7 modifications. These same three nucleotides, conserved in the wild-type RRE, were also found to be critical for Rev binding in a modification interference study using short RNAs containing the RRE IIB stem loop (Kjems et al. (1992) EMBO J. 11(3):1119-1129). The second region centers around G10 -A11 -G12 with interference again from phosphate, base-pairing face, and N7 modifications. Additionally, there is a smaller "mini-region" encompassing the stretch C6 -A7 -U8, with phosphate and base-pairing face modifications interfering with binding.
Throughout the ligand, many base-pairing face modifications showed binding interference, most likely because of perturbations in the ligand's secondary structure. Two of the "bulge" bases, U9 and A14, did not exhibit modification interference, indicating that both have neither a role in specific base-pairing interactions/stacking nor in contacting the protein.
Chemical modification protection when RNA is bound to Rev. The "footprinting" chemical modification data is summarized in FIG. 20. Four positions U8, A13, A15, and A27, showed at least two-fold reduction in modification of base-pairing faces (and a like reduction in N7 modification for the A positions) while bound to Rev protein. The slight N7 modifications of G10 -A11 -G12 under native conditions were not detected when the ligand was modified in the presence of Rev. G32, unmodified in chemical probing of the RNA native structure, shows strong modification of its base-pairing face and the N7 position when complexed with Rev. U31 and U33, 5' and 3' of G32, show slight CMCT modification when the ligand is bound to protein.
Secondary SELEX using biased randomization of template. A template was synthesized as shown in FIG. 21 in which the Rev ligand 6a sequence was mixed with the other three nucleotides at each position in the ratio of 62.5 (for the 6a sequence) to 12.5 for each of the other three nucleotides. This biased template gave rise to RNAs with background affinity for Rev protein (Kd=10-7). Six rounds of SELEX yielded the list of sequences shown in FIG. 21. The frequency distribution of the nucleotides and base pairs found at each position as it differs from that expected from the input distribution during template synthesis is shown in FIGS. 22 and 23. A new consensus based on these data is shown in FIG. 24. The most significant differences from the sequence of Rev ligand 6a are replacement of the relatively weak base pair A7-U31 with a G-C pair and allowed or prefered substitution of U9 with C, A14 with U, U22 with G. Absolutely conserved positions are at sites G10, A11, G12; A15, C16, A17; U24, G25; and C28, U29, C30. No bases were found substituted for G26 and A25, although there was one and three deletions found at those positions respectively. Two labeled transcripts were synthesized, one with a simple ligand 6a-like sequence, and one with substitutions by the significant preferences found in FIG. 24. These RNAs bound identically to Rev protein.
Most of the substitutions in the stem region increase its stability. There does not seem to be significant selection of stems of length longer than 5 base-pairs although this could be a selection for replicability (for ease of replication during the reverse transcription step of SELEX, for example). There is some scattered substitution of other nucleotides for U9 in the original SELEX reported in U.S. patent application Ser. No. 07/714,131 filed Jun. 10, 1991, but this experiment shows prefered substitution with C. Deletions of A27 also appeared in that original SELEX. A surprising result is the appearance of C18-A pairings in place of C18-G23 at a high frequency.
The reason there may be preferences found in this experiment that do not improve measured binding affinity may lie in the differences in the binding reactions of SELEX and these binding assays. In SELEX a relatively concentrated pool of heterogeneous RNA sequences (flanked by the requisite fixed sequences) are bound to the protein. In binding assays low concentrations of homogeneous RNA sequence are bound. In SELEX there may be selection for more discriminating conformational certainty due to the increased probability of intermolecular and intramolecular contacts with other RNA sequences. In the therapeutic delivery of concentrated doses of RNA ligands and their modified homologs, these preferences found in secondary SELEXes may be relevant.
EXAMPLE I: ELUCIDATION OF IMPROVED NUCLEIC ACID LIGAND SOLUTION FOR HIV-RT
RNA synthesis. In vitro transcription with oligonucleotide templates was conducted as described by Milligan et al. (1988). All synthetic nucleic acids were made on an Applied Biosystems model 394-08 DNA/RNA synthesizer using standard protocols. Deoxyribonucleotide phosphoramidites and DNA synthesis solvents and reagents were purchased from Applied Biosystems. Ribonucleotide and 2'-methoxy-ribonucleotide phosphoramidites were purchased from Glen Research Corporation. For mixed base positions, 0.1M phosphoramidite solutions were mixed by volume to the proportions indicated. Base deprotection was carried out at 55° C. for 6 hours in 3:1 ammonium hydroxide:ethanol. t-butyl-dimethylsilyl protecting groups were removed from the 2'-OH groups of synthetic RNAs by overnight treatment in tetrabutylammonium fluoride. The deprotected RNAs were then phenol extracted, ethanol precipitated and purified by gel electrophoresis.
Affinity assays with labeled RNA and HIV-RT. Model RNAs for refinement of the 5' and 3' boundaries and for determination of the effect of substitutions were labeled during transcription with T7 RNA polymerase as described in Tuerk et al. (1990) except that a-32P-ATP was used, in reactions of 0.5 mM C,G, and UTP with 0.05 mM ATP. Synthetic oligonucleotides and phosphatased transcripts (as in Tuerk et al., 1990) were kinased as described in Gauss et al. (1987). All RNA-protein binding reactions were done in a "binding buffer" of 200 mM KOAc, 50 mM Tris-HCl pH 7.7, 10 mM dithiothreitol with exceptions noted for chemical protection experiments below. RNA and protein dilutions were mixed and stored on ice for 30 minutes then transferred to 37° C. for 5 minutes. In binding assays the reaction volume was 60 ul of which 50 ul was assayed. Each reaction was suctioned through a pre-wet (with binding buffer) nitrocellulose filter and rinsed with 3 mls of binding buffer after which it was dried and counted for assays or subjected to elution and assayed for chemical modification. In comparisons of binding affinity, results were plotted and the protein concentration at which half-maximal binding occurred (the approximate Kd in conditions of protein excess) was determined graphically.
Selection of modified RNAs by HIV-RT. Binding reactions were as above except that rather than to vary the amount of HIV-RT added to a reaction, the volume of reaction was increased in order to lower concentration. RNAs that were modified under denaturing conditions were selected at concentrations of 20, 4 and 0.8 nanomolar HIV-RT (in volumes of 1, 5 and 25 mls of binding buffer.) The amount of RNA added to each reaction was equivalent for each experiment (approximately 1-5 picomoles). RNA was eluted from filters as described in Tuerk et al. (1990) and assayed for modified positions. In each experiment a control was included in which unselected RNA was spotted on a filter, eluted and assayed for modified positions in parallel with the selected RNAs. Determinations of variation in chemical modification for selected versus unselected RNAs were made by visual inspection of exposed films of electrophoresed assay products with the following exceptions. The extent of modification interference by ENU was determined by densitometric scanning of films using an LKB laser densitomer. An index of modification interference (M.I.) at each position was calculated as follows:
M.I.=(O.D. unselected/O.D. unselected
A20) / (O.D. selected/O.D. selected A20)
where the value at each position assayed for selected modified RNA (O.D.selected) is divided by that value for position A20 (O.D.selected A20) and divided into likewise normalized values for the unselected lane. All values of M.I. greater than 2.0 are reported as interfering and greater than 4.0 as strongly interfering. In determination of the effects of mixed substitution of 2'-methoxys for 2' hydroxyls (on the ribose at each nucleotide position) gels of electrophoresed hydrolysis products were counted on an Ambis detection system directly. The counts associated with each band within a lane were normalized as shown above but for position A17. In addition, determinations were done by laser densitometry as described below.
Chemical modification of RNA. A useful review of the types of chemical modifications of RNA and their specificities and methods of assay was done by Ehresmann et al. (1987). Modification of RNA under native conditions was done at 200 mM KOAc, 50 mM Tris-HCl pH 7.7 at 37° C. with ethylnitrosourea (ENU) (1/5 dilution v/v of room temperature ENU-saturated ethanol) for 1-3 hours, dimethyl sulfate (DMS) (1/750-fold dilution v/v) for eight minutes, kethoxal (0.5 mg/ml) for eight minutes, carbodiimide (CMCT) (8 mg/ml) for 20 minutes, and diethyl pyrocarbonate (DEPC) (1/10 dilution v/v for native conditions or 1/100 dilution for denaturing conditions) for 45 minutes, and under the same conditions bound to HIV-RT with the addition of 1 mM DTT. The concentrations of modifying chemical reagent were identical for denaturing conditions (except where noted for DEPC); those conditions were 7M urea, 50 mM Tris-HCl pH 7.7, 1 mM EDTA at 90° C. for 1-5 minutes except during modification with ENU which was done in the absence of 7M urea.
Assay of chemical modification. Positions of chemical modification were assayed by reverse transcription for DMS, kethoxal and CMCT on the lengthened ligand B RNA, 5'-GGUCCGAAGUGCAACGGGAAAAUGCACUAUGAAAGAAU-UUUAUAUCUCUAUUG AAAC-3' (SEQ ID NO: 4) (the ligand B sequence is underlined), to which is annealed the oligonucleotide primer 5'-CCGGATCCGTTTCAATAGAG-ATATAAAATTC-3'(SEQ ID NO:5) ; reverse transcription products (obtained as in Gauss et al., 1987) were separated by electrophoresis on 10% polyacrylamide gels. Positions of ENU and DEPC modification were assayed as in Vlassov et al. (1980) and Peattie and Gilbert (1980) respectively (separated by electrophoresis on 20% polyacrylamide gels). Assay of 2'-methoxy ribose versus ribose at various positions was assayed by alkaline hydrolysis for 45 minutes at 90° C. in 50 mM sodium carbonate pH 9.0.
Modification of RNA in the presence of HIV-RT. Conditions were as for modification of native RNA. Concentrations of HIV-RT were approximately 10-fold excess over RNA concentration. In general protein concentrations ranged from 50 nM to 1 uM.
SELEX isolation of accessory contacts with HIV-RT. The starting RNA was transcribed from PCRd templates synthesized from the following oligonucleotides:
5'-GGGCAAGCTTTAATACGACTCACTATAGGTCCGAAGTGCAACGGGAAAATG-CA CT-3' (5' primer) (SEQ ID NO: 6),
5'-GTTTCAATAGAGATATAAAATTCTTTCATAG-3 ' (3 ' primer)(SEQ ID NO: 7),
5'-GTTTCAATAGAGATATAAAATTCTTTCATAG-[30N]AGTGCATTTTCCCGTTG C-ACTTCGGACC-3' (variable template) (SEQ ID NO: 8).
SELEX was performed as described previously with HIV-RT with the following exceptions. The concentration of HIV-RT in the binding reaction of the first SELEX round was 13 nanomolar, RNA at 10 micromolar, in 4 mls of binding buffer, in the rounds 2 through 9 selection was done with 2.6 nanomolar HIV-RT, 1.8 micromolar RNA in 20 mls of buffer, in rounds 10-14 we used 1 nanomolar HIV-RT, 0.7 micromolar RNA in 50 mls, and for rounds 15 and 16 we used 0.5 nanomolar HIV-RT, 0.7 micromolar RNA in 50 mls of binding buffer.
REFERENCES TO EXAMPLE I
Ehresman, C., Baudin, F., Mougel, M., Romby, P., Ebel, J-P. Ehresman, B. (1987) Probing the structure of RNAs in solution. Nuc. Acids. Res. 15:9109-9128.
Freemont, P. S., Friedman, J. M., Beese, M. R., Sanderson, M. R. and Steitz, T. A. (1988) Proc. Natl. Acad. Sci. USA 85:8924.
Freier, S. M., Kierzed, R., Jaeger, J. A., Suigimoto, N., Caruthers, M. H., Neilson, T., and Turner, D. H. (1986) Proc. Natl. Acad. Sci. USA 83:9373-9377.
Gauss, P., Gayle, M., Winter, R. B., Gold, L. (1987) Mol. Gen. Genet. 206:24.
Kohlstaedt, L. A., Wang, J., Friedman, J. M., Rice, P. A. and Steitz, T. A. (1992) Crystal structure at 3.5 Å resolution of HIV-1 reverse transcriptase complexed with an inhibitor. Science 256:1783-1790.
Milligan, J. F., Groebe, D. R., Witherell, G. W. and Uhlenbeck, O. C. (1987) Oligoribonucleotide synthesis using T7 RNA polymerase and synthetic DNA templates. Nucleic Acids Res. 15:8783-8798.
Moazed, D., Stern, S. and Noller, H. (1986) Rapid chemical probing of conformation in 16S ribosomal RNA and 30S ribosomal subunits using primer extension. J. Mol. Biol. 187:399-416.
Peattie, D. and Gilbert, W. (1980) Chemical probes for higher order structure in RNA. Proc. Natl. Acad. Sci. USA 77:4679-4682.
Peattie, D. and Herr, W. (1981) Chemical probing of the tRNA-ribosome complex. Proc. Natl. Acad. Sci. USA 78:2273-2277.
Roald, M. A., Perona, J., So/ ll, D. and Steitz, T. A. (1989) Science 246:1135.
Tuerk, C., Eddy, S., Parma, D. and Gold, L. (1990) The translational operator of bacteriophage T4 DNA polymerase. J. Mol. Biol. 213:749.
Tuerk, C. and Gold, L. (1990) Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249:505-510.
Tuerk, C., MacDougal, S. and Gold, L. (1992) RNA pseudoknots that inhibit HIV-1 reverse transcriptase. Proc. Natl. Acad. Sci. USA 89:6988-6992.
Vlassov, V., Giege, R. and Ebel, J. -P. (1980) The tertiary structure of yeast tRNAPhe in solution studied by phosphodiester bond modification with ethylnitrosourea. FEBS 120:12-16.
EXAMPLE II: ELUCIDATION OF IMPROVED NUCLEIC ACID LIGAND SOLUTIONS FOR HIV-1 REV PROTEIN
The Rev ligand sequence used for chemical modification is shown in FIG. 12 (the numbering scheme shown will be used hereinafter). RNA for modification was obtained from T7 RNA polymerase transcription of synthetic oligonucleotide templates. ENU modification was carried out on the ligand sequence as shown in FIG. 12. DMS, kethoxal, CMCT, and DEPC modifications were carried out on a extended ligand sequence, and analyzed by reverse transcription with the synthetic oligonucleotide primer shown in FIG. 12.
Chemical Modification of RNA
Chemical modification techniques for nucleic acids are described in general in Ehresmann et al. (1987) Nuc. Acids. Res. 15:9109-9128. Modification of RNA under native conditions was performed in 200 mM KOAc, 50 mM Tris-HCl pH 7.7, 1 mM EDTA at 37 ° C. Modification under denaturing conditions was done in 7M urea, 50 mM Tris-HCl pH 7.7 at 90 ° C. Concentration of modifying agents and incubation times are as follows: ethylnitrosourea (ENU)-1/5 dilution v/v of ethanol saturated with ENU, native 1-3 hours, denaturing 5 minutes; dimethyl sulfate (DMS)-1/750-fold dilution v/v, native 8 minutes, denaturing 1 minute; kethoxal--0.5 mg/ml, native 5 minutes, denaturing 2 minutes; carbodiimide (CMCT)--10 mg/ml, native 30 minutes, denaturing 3 minutes; diethyl pyrocarbonate (DEPC)--1/10 dilution v/v, native 10 minutes, denaturing 1 minute.
Modification Interference of Rev Binding
RNAs chemically modified under denaturing conditions were selected for Rev binding through filter partitioning. Selections were carried out at Rev concentrations of 30, 6, and 1.2 nanomolar (in respective volumes of 1, 5, and 25 mls of binding buffer; 200 mM KOAc, 50 mM Tris-HCl pH 7.7, and 10 mM dithiothreitol), Approximately 3 picomoles of modified RNA were added to each protein solution, mixed and stored on ice for 15 minutes, and then transferred to 37° C. for 10 minutes. Binding solutions were passed through pre-wet nitrocellulose filters, and rinsed with 5 mls of binding buffer. RNA was eluted from the filters as described in Tuerk et al. (1990) Science 24:505-510 and assayed for modified positions that remained. Modified RNA was also spotted on filters and eluted to check for uniform recovery of modified RNA.
The extent of modification interference was determined by densitometric scanning of autoradiographs using LKB (ENU) and Molecular Dynamics (DMS, kethoxal, CMCT, and DEPC) laser densitometers. Values for modified phosphates and bases were normalized to a chosen modified position for both selected and unselected lanes; the values for the modified positions in the selected lane were then divided by the corresponding positions in the unselected lane (for specific normalizing positions see FIGS. 15-19). Values above 4.0 for modified bases and phosphates are designated as strongly interfering, and values above 2.0 are termed slightly interfering.
Modification of RNA in the Presence of Rev
"Footprinting" of the Rev ligand, modification of the RNA ligand in the presence of Rev protein, was performed in 200 mM KOAc, 50 mM Tris-Cl pH 7.7, 1 mM DTT, and 5 mM MgCl. Concentration of protein was 500 nanomolar, and approximately in 3-fold molar excess over RNA concentration. Modification with protein present was attempted with all modifying agents listed above except ethylnitrosourea (ENU).
Assay of Chemically Modified RNA
Positions of ENU modification were detected as in Vlassov et al. (1980) FEBS 120:12-16 and separated by electrophoresis on 20% denaturing acrylamide gels. DMS, kethoxal, CMCT, and DEPC were assayed by reverse transcription of the extended Rev ligand with a radiolabelled oligonucleotide primer (FIG. 12) and separated by electrophoresis on 8% denaturing acrylamide gels.
SELEX with Biased Randomization
The templates for in vitro transcription were prepared by PCR from the following oligonucleotides:
5'-CCCGGATCCTCTTTACCTCTGTGTGagatacagagtccacaaacgtgttc tcaatgcacccGGTCGGAAGGCCATCAATAGTCCC-3' (template oligo) (SEQ ID NO: 9)
5'-CCGAAGCTTAATACGACTCACTATAGGGACTATTGATGGCCTTCCGACC-3'(5' primer) (SEQ ID NO: 10)
5'-CCCGGATCCTCTTTACCTCTGTGTG-3' (3' primer) (SEQ ID NO: 11)
where the small case letters in the template oligo indicates that at each position that a mixture of reagents were used in synthesis by an amount of 62.5% of the small case letter, and 12.5% each of the other three nucleotides.
SELEX was conducted as described previously with the following exceptions. The concentration of HIV-1 Rev protein in the binding reactions of the first and second rounds was 7.2 nanomolar and the RNA 4 micromolar in a volume of 10 mls (of 200 mM potassium acetage, 50 mM Tris-HCl pH 7.7, 10 mM DTT). For rounds three through six the concentration of Rev protein was 1 nanomolar and the RNA 1 micromolar in 40 mls volume. HIV-1 Rev protein was purchased from American Biotechnologies, Inc.
__________________________________________________________________________
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(iii) NUMBER OF SEQUENCES: 83
(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
UUCCGNNNNNN NNCGGGAAAA21
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
UCCGNNNNNNNNCGGGAAAANNNN 24
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
NNUUCCGNNNNNNNNCGGGAAAANNNN 27
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 57 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
GGUCCGAAGUGCAACGGGAAAAUGCACUAUGAAAGAAUUUUAUAUCUCUA5 0
UUGAAAC57
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 31 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
CCGGATCCGT TTCAATAGAGATATAAAATTC31
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 55 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
GGGCAAGCTTTAATACGACTCACTA TAGGTCCGAAGTGCAACGGGAAAAT50
GCACT55
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 31 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
GTTTCAATAGAGATATAAAATTCTTTCATAG31
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
GTTTCAATAGAGATATAAAATTCTTTCATAGNNNNNNNNNNNNNNNNNNN50
NNNNNNNNNNNAGTGCATTTTCCCGTTGCACTTCGGACC89
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 85 base pairs
(B) TYPE: nucleic acid
 (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:
CCCGGATCCTCTTTACCTCTGTGTGAGATACAGAGTCCACAAACGTGTTC50
TCAATGCACCCGGTCGGAAGGCCATCAATAGTCCC85
(2) INFORMATION FOR SEQ ID NO:10:
 (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 49 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:
CCGAAGCTTAATACGACTCACTATAGGGACTATTGATGGCCTTCCGACC49
(2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 25 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:
CCCGGATCCTCTTTACCTCTGTGTG25
(2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
 (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ix) FEATURE:
OTHER INFORMATION: N's at positions 9-11 are base
paired with N's at positions
21-23
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
UCCSANNNNNNASGGGANAANNN 23
(2) INFORMATION FOR SEQ ID NO:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 59 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(iii) MOLECULE TYPE: RNA
(ix) FEATURE:
(D) OTHER INFORMATION: N at position 29 is 6-8 bases
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
GGUCCGAA GUGCAACGGGGAAAAUGCACNNNNNNNNNNNNNNNNNNNNNN50
NNNNNNNNN59
(2) INFORMATION FOR SEQ ID NO:14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
GGUCCGAAGUGCAACGGGAAAAUGCACUAGCUCGUGAGGCUUUCGUGCUG50
UUCCGAGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 87 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:
GGUCCGAAGUGCAACGGGAAAAUGCACUGCAUGUGAGGCGGUAACGCUGU50
UCCGUGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC 87
(2) INFORMATION FOR SEQ ID NO:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:
GGUCCGAAGUGCAACGGGAAAAUGCACUGGUGAGUGAGGCCGAUGCUGUU50
CCUCGCCGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 91 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:
GGUCCGAAGUG CAACGGGAAAAUGCACUGACGCGCGAGGUCUUGGUACUG50
UUCCGUGGCUCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC91
(2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
GGUCCGAAGUGCAACGGGAAAAUGCACUCUGGGUGAGACUUGAAGUCGUU50
CCCCAGGUCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:19:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19:
GGUCCGAAGUGCAACGGGAAAAUGCACUCCCGGUGAAGCAUAAUGCUGUU50
CCUGGGGUCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC 89
(2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20:
GGUCCGAAGUGCAACGGGAAAAUGCACUGGGAGUGAGGUUCCCCGUUCCU50
 CCCGCACCCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:21:
GGUCCGAAGUGCAAC GGGAAAAUGCACUAGCGAUGUGAAGUGAUACUGGU50
CCAUCGUGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:
GGUCCGAAGUGCAACGGGAAAAUGCACUCACAGUGAGCCUUCUGGUGGUC50
CUGUGUGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:23:
(i) SEQUENCE CHARACTERISTICS:
( A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:23:
GGUCCGAAGUGCAACGGGAAAAUGCACUUGUUGUGAGUGGUUGAUUCCAU50
GGUCCAACCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC 89
(2) INFORMATION FOR SEQ ID NO:24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:
GGUCCGAAGUGCAACGGGAAAAUGCACUGCCUGUGAGCUGUUUAGCGGUC50
CAGG UCGUCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:
GGUCCGAAGUGCAACGGGA AAAUGCACUCAAGGCGAAGACUUAGUCUGCU50
CCCUGUGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:26:
GGUCCGAAGUGCAACGGGAAAAUGCACUUGCGUCGAAGUUAAUUCUGGUC50
GAUGCCACUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:27:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 90 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:27:
GGUCCGAAGUGCAACGGGAAAAUGCACUUUCAAUGAGGUAUGUAAUGAUG50
GUCGUGCGCCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC 90
(2) INFORMATION FOR SEQ ID NO:28:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:28:
GGUCCGAAGUGCAACGGGAAAAUGCACUGCGGGAGAGUCUUUUGACGUUG50
CUCCUGCG CUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:29:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:29:
GGUCCGAAGUGCAACGGGAAAA UGCACUCAUGGGAGCCCAUCGAUUCUGG50
GUGUUGCCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:30:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
( D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:30:
GGUCCGAAGUGCAACGGGAAAAUGCACUUGCACAGAGCCAAAUUUGGUGU50
UGCUGUGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
 (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:31:
GGUCCGAAGUGCAACGGGAAAAUGCACUGGCCAGAGCUUAAAUUCAAGUG50
UUGCUGGCCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:32:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:32:
GGUCCGAAGUGCAACGGGAAAAUGCACUCAUAGCAGUCCUUGAUACUAUG50
GAUGGUGGCUA UGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:33:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:33:
GGUCCGAAGUGCAACGGGAAAAUGCA CUGGAUGCAAGUUAACUCUGGUGG50
CAUCCGUCCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:34:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 88 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:34:
GGUCCGAAGUGCAACGGGAAAAUGCACUCAGUGGAGAUUAAGCCUCGCUA50
GGGGCCGCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC88
(2) INFORMATION FOR SEQ ID NO:35:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 base pairs
 (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ix) FEATURE:
(D) OTHER INFORMATION: Y at position 7 is either U or
(ix) FEATURE:
(D) OTHER INFORMATION: S at position 1 is base paired
to the S at position 34
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:35:
SUGCGUYGAG AUACACNNNGGUGGACUCCCGCAS34
(2) INFORMATION FOR SEQ ID NO:36:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 26 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: RNA
(ix) FEATURE:
(D) OTHER INFORMATION: The N's as positions 6-11 are
base paired to the N's at
positions 21- 26
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:36:
UCCGANNNNNNACGGGANAANNNNNN26
(2) INFORMATION FOR SEQ ID NO:37:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 89 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: RNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:37:
GGUCCGAAGUGCAACGGGAAAAUGCACUNNNNNNNNNNNNNNNNNNNNNN50
NNNNNNNNCUAUGAAAGAAUUUUAUAUCUCUAUUGAAAC89
(2) INFORMATION FOR SEQ ID NO:38:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 51 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: RNA
(ix) FEATURE:
(D) OTHER INFORMATION: N's at postiions 29-32 are
based paired with N's at
positions 48- 51
 (ix) FEATURE:
(D) OTHER INFORMATION: N's at postiions 38-39 are
based paired with N's at
positions 40- 41
(ix) FEATURE:
(D) OTHER INFORMATION: N at position 44 is either G
or U
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:38:
GGUCCGAAGUGCAACGGGAAAAUGCACUNNNNGUGARNN NNUGNUCCNNN50
N51
(2) INFORMATION FOR SEQ ID NO:39:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 54 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: RNA
(ix) FEATURE:
(D) OTHER INFORMATION: N's at postiions 29-34 are
based paired with N's at
positions 48- 53
(ix) FEATURE:
(D) OTHER INFORMATION: N's at postiions 37-39 are
based paired with N's at
positions 40- 42
(ix) FEATURE:
(D) OTHER INFORMATION: N at position 45 is either G
or U
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:39:
GGUCCGAAGUGCAACGGGAAAAUGCACUNNNNNNAGNNNNNNUGNUGNNN50
NNNN54
(2) INFORMATION FOR SEQ ID NO:40:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 37 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:40:
GGGUGCAUUGAGAAACACGUUUGUGGACUCUGUAUCU37
(2) INFORMATION FOR SEQ ID NO:41:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 66 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:41:
GGGUGCAUUGAGAAACACGUUUGUGGACUCUGUAUCUAUGAAAGAAUUUU50
AUAUCUCUAUUGAAAC 66
(2) INFORMATION FOR SEQ ID NO:42:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:42:
GTTTCAATAGAGATATAAAATTC23
(2) INFORMATION FOR SEQ ID NO:43:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:43:
GGGACUAUUGAUGGCCUUCCGACCNNNNNNNNNNNNNNNNNNNNNNNNNN50
NNNNNNNNNNNCACACAGAGGUA AAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:44:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:44:
GGGACUAUUGAUGGCCUUCCGACCGGGUGCAUUGAGAA ACACGUUUGUGG50
ACUCUGUAUCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:45:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 84 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:45:
GGGACUAUUGAUGGCCUUCCGACCUGGUGCGUUGAGAAACAGGUUUUUGG50
ACUCCGUACCACACAGAGGUAAAGAGGAUCCGGG84
(2) INFORMATION FOR SEQ ID NO:46:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:46:
GGGACUAUUGAUGGCCUUCCGACCGUAUGCAUUGAGAGACACACUUGUGG50
ACUCUGCAUCCCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:47:
 (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:47:
GGGACUAUUGAUGGCCUUCCGACCAGAUGGAUUGAGAAACACUAUUAUGG50
ACUCUCCAUCGCACACAGAGGUAAAGA GGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:48:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:48:
GGGACUAUUGAUGGCCUUCCGACCAGCUUCGUCGAGAUACA CGUUGAUGG50
ACUCCGAAGCACACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:49:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:49:
GGGACUAUUGAUGGCCUUCCGACCUCGUACGUUGAGAAACAAGUUUAUGG50
ACUCCGUACCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:50:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
 (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:50:
GGGACUAUUGAUGGCCUUCCGACCUCGAUCGUUGAGAUACACGCUAGUGG50
ACUCCGAAACUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:51:
 (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:51:
GGGACUAUUGAUGGCCUUCCGACCUACUGCAUCGAGAUACACGUUUGUGG50
ACUCUGCACAUCACACAGAGGUAAAGAGGA UCCGGG86
(2) INFORMATION FOR SEQ ID NO:52:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:52:
GGGACUAUUGAUGGCCUUCCGACCUGAUACGUUGAGAAACACAAU GCUGG50
ACUCCGCAUCCCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:53:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:53:
 GGGACUAUUGAUGGCCUUCCGACCGCCUGCAUUGAGAAACAGGAUUCUGG50
ACUCUGCCACUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:54:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
 (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:54:
GGGACUAUUGAUGGCCUUCCGACCCGCUAUGUUGAGAAACACUUUGCUGG50
ACUCCGUAGCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:55:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 85 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:55:
GGGACUAUUGAUGGCCUUCCGACCUACUGCAUCGAGAAACACGUAAGUGA50
CUCUGCAUCCCACACAGAGGUAAAGAGGAUCCGG G85
(2) INFORMATION FOR SEQ ID NO:56:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:56:
GGGACUAUUGAUGGCCUUCCGACCCGGUACGUCGAGAUACACGAAGAUG G50
ACUCCGUAUCGCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:57:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:57:
GGGA CUAUUGAUGGCCUUCCGACCAACUCCAUCGAGAAACACGAUAGUGG50
ACUCUGGAGCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:58:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
 (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:58:
GGGACUAUUGAUGGCCUUCCGACCGGAGACGUCGAGAAACACGUUUGUGG50
ACUCCGUCUCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:59:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:59:
GGGACUAUUGAUGGCCUUCCGACCAGCUACAUCGAGAAACAAGAUUUUGG50
ACUCUGUAGCGCACACAGAGGUAAAGAGGAUCCGGG 86
(2) INFORMATION FOR SEQ ID NO:60:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 83 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:60:
GGGACUAUUGAUGGCCUUCCGACCAAGUGCAUUGAGAUACAAAUGAUUGG 50
ACUCUGCACACACAGAGGUAAAGAGGAUCCGGG83
(2) INFORMATION FOR SEQ ID NO:61:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:61:
GGGACUAU UGAUGGCCUUCCGACCUGCUACGUUGAGAUACACGUUGAUGC50
ACUCCGUAGCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:62:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:62:
GGGACUAUUGAUGGCCUUCCGACCAGCUACGUUGAGAUACACGUUACGUG50
GCUCCGUAGCCCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:63:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:63:
GGGACUAUUGAUGGCCUUCCGACCGAGUGGCUCGAGAAACAGGUUGCUGG50
ACUCGCCACAUCACACAGAGGUAAAGAGGAUCCGGG 86
(2) INFORMATION FOR SEQ ID NO:64:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:64:
GGGACUAUUGAUGGCCUUCCGACCUCGUGCGUCGAGCAACACGUUGAUGG50
ACUCCGCACAGCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:65:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:65:
GGGACUAUUGA UGGCCUUCCGACCGGCACCGUUGAGAAACACAUGCGUGG50
ACUCCGUGCCCCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:66:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:66:
GGGACUAUUGAUGGCCUUCCGACCUCCUGCAUUGAGAAACAGUGAUCUGG50
ACUCUGCAACUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:67:
(i) SEQUENCE CHARACTERISTICS:
 (A) LENGTH: 85 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:67:
GGGACUAUUGAUGGCCUUCCGACCUGUGGAUUGAGCAACACGUGAGUGGA50
CUCUCCACAUCACACAGAGGUAAAGAGGAUCCGGG 85
(2) INFORMATION FOR SEQ ID NO:68:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:68:
GGGACUAUUGAUGGCCUUCCGACCCCGUGCGUUGAGACACACACCGAUGG50
 ACUCCGCAUGUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:69:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:69:
GGGACUAUUGAUGGC CUUCCGACCAGCUGCAUCGAGAUACACGAUUGUGG50
ACUCUGCAGCCCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:70:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 87 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:70:
GGGACUAUUGAUGGCCUUCCGACCAGAUUCGUUGAGAAACACAUGGGUGG50
ACUCUCCCGCUACACACAGAGGUAAAGAGGAUCCGGG87
(2) INFORMATION FOR SEQ ID NO:71:
(i) SEQUENCE CHARACTERISTICS:
( A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:71:
GGGACUAUUGAUGGCCUUCCGACCAGAUGGAUUGAGAAACACGUUCGUGG50
ACUCUCCAACUCACACAGAGGUAAAGAGGAUCCGGG 86
(2) INFORMATION FOR SEQ ID NO:72:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:72:
GGGACUAUUGAUGGCCUUCCGACCGACUGCAUCGAGAAACACUGAUGUGG50
CCUC CGCACGGCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:73:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:73:
GGGACUAUUGAUGGCCUUC CGACCAGCUACGUUGAGAAACAGUAUAAUGG50
ACUCCGUAGCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:74:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
 (D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:74:
GGGACUAUUGAUGGCCUUCCGACCGAGUGCGUCGAGAAACACAUUUGUGG50
ACUCCGCACACCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:75:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:75:
GGGACUAUUGAUGGCCUUCCGACCUCGUACGUUGAGAAACACGCUAGUGG50
ACUCCGUAUGUCACACAGAGGUAAAGAGGAUCCGGG 86
(2) INFORMATION FOR SEQ ID NO:76:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:76:
GGGACUAUUGAUGGCCUUCCGACCAGAUACGUUGAGAGACACGCACGUGG50
ACUCCGUA UCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:77:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:77:
GGGACUAUUGAUGGCCUUCCGA CCAGGAUCACAGAGAAACACCGUGGGUG50
GCUCCCUCUAUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:78:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 87 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
( D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:78:
GGGACUAUUGAUGGCCUUCCGACCGUGCGCAUCGAGAAACACGUUGAUGG50
ACUCUGCAUGCACACACAGAGGUAAAGAGGAUCCGGG87
(2) INFORMATION FOR SEQ ID NO:79:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
 (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:79:
GGGACUAUUGAUGGCCUUCCGACCGAGAGGAUCGAGAAACACGUAUGUGG50
ACUCUCCAUCUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:80:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:80:
GGGACUAUUGAUGGCCUUCCGACCGGAUGGAUUGAGACACACGUAUGUGG50
ACUCUCCAUCA CACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:81:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:81:
GGGACUAUUGAUGGCCUUCCGACCUC GGGCAUUGAGAUACACGUAGAUGG50
ACUCUGUCUCACACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:82:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 86 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:82:
GGGACUAUUGAUGGCCUUCCGACCUGGACCGUAGAGAAACACGUUUGAUG50
GCUCCCUCUGUCACACAGAGGUAAAGAGGAUCCGGG86
(2) INFORMATION FOR SEQ ID NO:83:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs
 (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: RNA
(ix) FEATURE:
(D) OTHER INFORMATION: N's at positions 1-5 are
paired with N's at positions
25-29
(ix) FEATURE:
(D) OTHER INFORMATION: N at position 16 is paired
with N at position 17
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:83:
NNNNNUUGAGAUACANNUGGACUCNNNNN29

Claims (10)

We claim:
1. A non-naturally occurring nucleic acid ligand to HIV reverse transcriptase (RT) obtained according to a method for identifying a nucleic acid ligand from a candidate mixture of nucleic acids, said method comprising:
a) contacting the candidate mixture with HIV-RT, wherein nucleic acids having an increased affinity to HIV-RT relative to the candidate mixture nucleic acids may be partitioned from the remainder of the candidate mixture;
b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; and
c) amplifying the increased affinity nucleic acids, in vitro, to yield a ligand-enriched mixture of nucleic acids, whereby nucleic acid ligands of HIV-RT may be identified and obtained nucleic acids.
2. The nucleic acid ligand to HIV-RT obtained by the method of claim 1 wherein said method further comprises:
d) preparing a modified nucleic acid that is identical to the nucleic acid ligand except for a single residue substitution; and
e) assessing the binding affinity of the modified nucleic acid relative to the nucleic acid ligand whereby nucleic acid ligands to HIV-RT may be obtained.
3. The nucleic acid ligand to HIV-RT may be obtained by the method of claim 1 wherein said method further comprises:
d) preparing a modified nucleic acid that is identical to the nucleic acid ligand except for the absence of one or more terminal residues; and
e) assessing the binding affinity of the modified nucleic acid relative to the nucleic acid ligand whereby nucleic acid ligands to HIV-RT may be obtained.
4. A non-naturally occurring nucleic acid ligand to HIV-RT having the RNA sequence (SEQ ID NO:12): ##STR1## wherein X=any base, and X-X' indicates a preferred base-pair.
5. A non-naturally occurring nucleic acid ligand to HIV-RT obtained according to a method for identifying an extended nucleic acid ligand from a candidate mixture of nucleic acids, said nucleic acid ligand being a ligand of HIV-RT comprising:
a) contacting the candidate mixture with HIV-RT, wherein nucleic acids having an increased affinity to HIV-RT relative to the candidate mixture nucleic acids may be partitioned from the remainder of the candidate mixture;
b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture;
c) amplifying the increased affinity nucleic acids, in vitro, to yield a ligand-enriched mixture of nucleic acids;
d) repeating steps a)-c), as necessary, to identify said nucleic acid ligand to HIV-RT;
e) contacting a second candidate mixture of nucleic acids with HIV-RT, wherein said second candidate mixture is comprised of nucleic acids having a fixed region and a randomized region, said fixed region corresponding to said nucleic acid ligand to HIV-RT identified in d), and wherein nucleic acids having an increased affinity to HIV-RT relative to the candidate mixture nucleic acids may be partitioned from the remainder of the candidate mixture;
f) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture;
g) amplifying the increased affinity nucleic acids to yield a ligand-enriched mixture of nucleic acids; and
h) repeating steps e)-h), as necessary, whereby said nucleic acid ligand to HIV-RT may be obtained.
6. A non-naturally occurring nucleic acid ligand obtained by the method of claim 5 having the RNA sequence (SEQ ID NO: 13): ##STR2## or the corresponding DNA sequence thereof; wherein x is any base, and Z is selected from the group consisting of the sequences set forth in FIG. 9 (SEQ ID NO:14-34).
7. A non-naturally occurring nucleic acid ligand to HIV-1 Rev obtained according to a method for identifying a nucleic acid ligand from a candidate mixture of nucleic acids, said method comprising:
a) contacting the candidate mixture with HIV-1 Rev, wherein nucleic acids having an increased affinity to HIV-1 Rev relative to the candidate mixture nucleic acids may be partitioned from the remainder of the candidate mixture;
b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; and
c) amplifying the increased affinity nucleic acids, in vitro, to yield a ligand-enriched mixture of nucleic acids, whereby nucleic acid ligands to HIV-1 Rev may be identified and obtained.
8. The nucleic acid ligand to HIV-1 Rev obtained by the method of claim 7 wherein said method further comprises:
d) preparing a modified nucleic acid that is identical to the nucleic acid ligand except for a single residue substitution; and
e) assessing the binding affinity of the modified nucleic acid relative to the nucleic acid ligand.
9. The nucleic acid ligand to HIV-1 Rev obtained by the method of claim 7 wherein said method further comprises:
d) preparing a modified nucleic acid that is identical to the nucleic acid ligand except for the absence of one or more terminal residues; and
e) assessing the binding affinity of the modified nucleic acid relative to the nucleic acid ligand whereby nucleic acid ligands to HIV-1 Rev may be obtained.
10. A non-naturally occurring nucleic acid ligand to HIV-1 Rev having a RNA sequence (SEQ ID NO:35): ##STR3## wherein X=any base. f5 whereby nucleic acid ligands to HIV-RT may be obtained
US07/964,624 1990-06-11 1992-10-21 Nucleic acid ligands to HIV-RT and HIV-1 rev Expired - Lifetime US5496938A (en)

Priority Applications (74)

Application Number Priority Date Filing Date Title
US07/964,624 US5496938A (en) 1990-06-11 1992-10-21 Nucleic acid ligands to HIV-RT and HIV-1 rev
AT93924904T ATE315101T1 (en) 1992-09-29 1993-09-28 NUCLEIC ACID LIGANDS AND THEIR PRODUCTION METHODS
CA002145761A CA2145761C (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for producing the same
DK93924904T DK0668931T3 (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for their preparation
JP6509298A JPH08501943A (en) 1992-09-29 1993-09-28 Nucleic acid ligand and method for producing the same
EP93924904A EP0668931B1 (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for producing the same
AU54411/94A AU689087B2 (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for producing the same
EP05023827A EP1683871B1 (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for producing the same
DE69333961T DE69333961T2 (en) 1992-09-29 1993-09-28 NUCLEIC ACID EQUIPMENT AND ITS MANUFACTURING METHODS
PT93924904T PT668931E (en) 1992-09-29 1993-09-28 NUCLEIC ACID LIGANDS AND METHODS FOR PRODUCING THEMSELVES
AT05023827T ATE518965T1 (en) 1992-09-29 1993-09-28 NUCLEIC ACID LIGANDS AND THEIR PRODUCTION METHODS
ES93924904T ES2257735T3 (en) 1992-09-29 1993-09-28 NUCLEIC ACID LINKS AND PRODUCTION PROCEDURE.
PCT/US1993/009296 WO1994008050A1 (en) 1992-09-29 1993-09-28 Nucleic acid ligands and methods for producing the same
US08/199,507 US5472841A (en) 1990-06-11 1994-02-22 Methods for identifying nucleic acid ligands of human neutrophil elastase
US08/233,012 US5849479A (en) 1990-06-11 1994-04-25 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US08/238,863 US5503978A (en) 1990-06-11 1994-05-06 Method for identification of high affinity DNA ligands of HIV-1 reverse transcriptase
US08/303,362 US5648214A (en) 1990-06-11 1994-09-09 High-affinity oligonucleotide ligands to the tachykinin substance P
US08/317,403 US5629155A (en) 1990-06-11 1994-10-03 High-affinity oligonucleotide ligands to immunoglobulin E (IgE)
US08/361,795 US5756287A (en) 1990-06-11 1994-12-21 High affinity HIV integrase inhibitors
US08/399,412 US5622828A (en) 1990-06-11 1995-03-06 High-affinity oligonucleotide ligands to secretory phospholipase A2 (sPLA2)
US08/430,709 US5660985A (en) 1990-06-11 1995-04-27 High affinity nucleic acid ligands containing modified nucleotides
US08/433,585 US5763566A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue SELEX
US08/433,126 US5688935A (en) 1990-06-11 1995-05-03 Nucleic acid ligands of tissue target
US08/434,425 US5789157A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/434,001 US5712375A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/433,124 US5750342A (en) 1990-06-11 1995-05-03 Nucleic acid ligands of tissue target
US08/437,667 US5864026A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/441,591 US5637682A (en) 1990-06-11 1995-05-16 High-affinity oligonucleotide ligands to the tachykinin substance P
US08/442,572 US5587468A (en) 1990-06-11 1995-05-16 High affinity nucleic acid ligands to HIV integrase
US08/442,062 US5595877A (en) 1990-06-11 1995-05-16 Methods of producing nucleic acid ligands
US08/441,828 US5734034A (en) 1990-06-11 1995-05-16 Nucleic acid ligand inhibitors of human neutrophil elastase
US08/443,407 US5786462A (en) 1990-06-11 1995-05-17 High affinity ssDNA ligands of HIV-1 reverse transcriptase
US08/447,172 US5726017A (en) 1990-06-11 1995-05-19 High affinity HIV-1 gag nucleic acid ligands
US08/447,169 US5811533A (en) 1990-06-11 1995-05-19 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US08/458,423 US5731144A (en) 1990-06-11 1995-06-02 High affinity TGFβ nucleic acid ligands
US08/458,424 US5731424A (en) 1990-06-11 1995-06-02 High affinity TGFβ nucleic acid ligands and inhibitors
US08/465,594 US5846713A (en) 1990-06-11 1995-06-05 High affinity HKGF nucleic acid ligands and inhibitors
US08/465,591 US5837834A (en) 1990-06-11 1995-06-05 High affinity HKGF nucleic acid ligands and inhibitors
US08/471,985 US5686592A (en) 1990-06-11 1995-06-06 High-affinity oligonucleotide ligands to immunoglobulin E (IgE)
US08/472,255 US5766853A (en) 1990-06-11 1995-06-07 Method for identification of high affinity nucleic acid ligands to selectins
US08/477,530 US5635615A (en) 1990-06-11 1995-06-07 High affinity HIV nucleocapsid nucleic acid ligands
US08/477,830 US5654151A (en) 1990-06-11 1995-06-07 High affinity HIV Nucleocapsid nucleic acid ligands
US08/479,725 US5674685A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/487,720 US5874557A (en) 1990-06-11 1995-06-07 Nucleic acid ligand inhibitors to DNA polymerases
US08/487,425 US5789163A (en) 1990-06-11 1995-06-07 Enzyme linked oligonucleotide assays (ELONAS)
US08/477,527 US5972599A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands of cytokines
US08/488,402 US5837456A (en) 1990-06-11 1995-06-07 High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US08/484,552 US5849890A (en) 1990-06-11 1995-06-07 High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US08/479,783 US5668264A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/484,557 US5693502A (en) 1990-06-11 1995-06-07 Nucleic acid ligand inhibitors to DNA polymerases
US08/472,256 US6001988A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands to lectins
US08/479,729 US5853984A (en) 1990-06-11 1995-06-07 Use of nucleic acid ligands in flow cytometry
US08/479,724 US5780228A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands to lectins
US08/481,710 US6028186A (en) 1991-06-10 1995-06-07 High affinity nucleic acid ligands of cytokines
US08/487,426 US5763173A (en) 1990-06-11 1995-06-07 Nucleic acid ligand inhibitors to DNA polymerases
US08/748,697 US5817785A (en) 1990-06-11 1996-11-13 Methods of producing nucleic acid ligands
US08/870,930 US6168778B1 (en) 1990-06-11 1997-06-06 Vascular endothelial growth factor (VEGF) Nucleic Acid Ligand Complexes
US08/918,304 US5958691A (en) 1990-06-11 1997-08-25 High affinity nucleic acid ligands containing modified nucleotides
US08/976,413 US6127119A (en) 1990-06-11 1997-11-21 Nucleic acid ligands of tissue target
US09/046,247 US6124449A (en) 1990-06-11 1998-03-23 High affinity TGFβ nucleic acid ligands and inhibitors
US09/165,616 US6344318B1 (en) 1990-06-11 1998-10-02 Methods of producing nucleic acid ligands
US09/364,902 US6232071B1 (en) 1990-06-11 1999-07-29 Tenascin-C nucleic acid ligands
US09/363,939 US6346611B1 (en) 1990-06-11 1999-07-29 High affinity TGfβ nucleic acid ligands and inhibitors
US09/407,234 US6184364B1 (en) 1990-06-11 1999-09-27 High affinity nucleic acid ligands containing modified nucleotides
US09/791,301 US6713616B2 (en) 1990-06-11 2001-02-23 High affinity TGFβ nucleic acid ligands and inhibitors
US09/854,662 US6596491B2 (en) 1990-06-11 2001-05-14 Tenascin-C nucleic acid ligands
US09/860,474 US6696252B2 (en) 1990-06-11 2001-05-18 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US10/040,497 US6716583B2 (en) 1990-06-11 2002-01-07 Methods of producing nucleic acid ligands
JP2003411496A JP2004097232A (en) 1992-09-29 2003-12-10 Nucleic acid ligand and production method thereof
US10/812,642 US20040258656A1 (en) 1990-06-11 2004-03-30 High affinity TGFbeta nucleic acid ligands and inhibitors
US10/818,954 US7368236B2 (en) 1990-06-11 2004-04-06 Methods of producing nucleic acid ligands
US11/286,221 US20060084797A1 (en) 1990-06-11 2005-11-23 High affinity TGFbeta nucleic acid ligands and inhibitors
JP2008134843A JP2008206524A (en) 1992-09-29 2008-05-22 Nucleic acid ligand and method for producing the same
JP2009177035A JP2009254380A (en) 1992-09-29 2009-07-29 Nucleic acid ligand and method for producing the same

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US53642890A 1990-06-11 1990-06-11
US07/714,131 US5475096A (en) 1990-06-11 1991-06-10 Nucleic acid ligands
US07/964,624 US5496938A (en) 1990-06-11 1992-10-21 Nucleic acid ligands to HIV-RT and HIV-1 rev

Related Parent Applications (4)

Application Number Title Priority Date Filing Date
US53642890A Continuation-In-Part 1990-06-11 1990-06-11
US07/714,131 Continuation-In-Part US5475096A (en) 1990-06-11 1991-06-10 Nucleic acid ligands
US07/931,473 Continuation-In-Part US5270163A (en) 1990-06-11 1992-08-17 Methods for identifying nucleic acid ligands
US08/234,997 Continuation-In-Part US5683867A (en) 1990-06-11 1994-04-28 Systematic evolution of ligands by exponential enrichment: blended SELEX

Related Child Applications (38)

Application Number Title Priority Date Filing Date
US53642890A Continuation-In-Part 1990-06-11 1990-06-11
US07/714,131 Continuation-In-Part US5475096A (en) 1990-06-11 1991-06-10 Nucleic acid ligands
US07/973,333 Continuation-In-Part US5476766A (en) 1990-06-11 1992-11-06 Ligands of thrombin
US11799193A Continuation-In-Part 1990-06-11 1993-09-08
US08/199,507 Continuation-In-Part US5472841A (en) 1990-06-11 1994-02-22 Methods for identifying nucleic acid ligands of human neutrophil elastase
US20551594A Continuation-In-Part 1990-06-11 1994-03-03
US08/233,012 Continuation-In-Part US5849479A (en) 1990-06-11 1994-04-25 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US08/238,863 Continuation-In-Part US5503978A (en) 1990-06-11 1994-05-06 Method for identification of high affinity DNA ligands of HIV-1 reverse transcriptase
US08/303,362 Continuation-In-Part US5648214A (en) 1990-06-11 1994-09-09 High-affinity oligonucleotide ligands to the tachykinin substance P
US08/317,403 Continuation-In-Part US5629155A (en) 1990-06-11 1994-10-03 High-affinity oligonucleotide ligands to immunoglobulin E (IgE)
US08/361,795 Continuation-In-Part US5756287A (en) 1990-06-11 1994-12-21 High affinity HIV integrase inhibitors
US08/434,425 Continuation-In-Part US5789157A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/437,667 Continuation-In-Part US5864026A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/433,126 Continuation-In-Part US5688935A (en) 1990-06-11 1995-05-03 Nucleic acid ligands of tissue target
US08/434,001 Continuation-In-Part US5712375A (en) 1990-06-11 1995-05-03 Systematic evolution of ligands by exponential enrichment: tissue selex
US08/442,062 Division US5595877A (en) 1990-06-11 1995-05-16 Methods of producing nucleic acid ligands
US08/447,169 Continuation-In-Part US5811533A (en) 1990-06-11 1995-05-19 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US08/458,423 Continuation-In-Part US5731144A (en) 1990-06-11 1995-06-02 High affinity TGFβ nucleic acid ligands
US08/458,424 Continuation-In-Part US5731424A (en) 1990-06-11 1995-06-02 High affinity TGFβ nucleic acid ligands and inhibitors
US08/465,594 Continuation-In-Part US5846713A (en) 1990-06-11 1995-06-05 High affinity HKGF nucleic acid ligands and inhibitors
US08/465,591 Continuation-In-Part US5837834A (en) 1990-06-11 1995-06-05 High affinity HKGF nucleic acid ligands and inhibitors
US08/488,402 Continuation-In-Part US5837456A (en) 1990-06-11 1995-06-07 High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US08/484,557 Continuation-In-Part US5693502A (en) 1990-06-11 1995-06-07 Nucleic acid ligand inhibitors to DNA polymerases
US08/479,725 Continuation-In-Part US5674685A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/472,256 Continuation-In-Part US6001988A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands to lectins
US08/477,530 Continuation-In-Part US5635615A (en) 1990-06-11 1995-06-07 High affinity HIV nucleocapsid nucleic acid ligands
US08/472,255 Continuation-In-Part US5766853A (en) 1990-06-11 1995-06-07 Method for identification of high affinity nucleic acid ligands to selectins
US08/481,710 Continuation-In-Part US6028186A (en) 1991-06-10 1995-06-07 High affinity nucleic acid ligands of cytokines
US08/477,830 Continuation-In-Part US5654151A (en) 1990-06-11 1995-06-07 High affinity HIV Nucleocapsid nucleic acid ligands
US08/477,527 Continuation-In-Part US5972599A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands of cytokines
US08/479,783 Continuation-In-Part US5668264A (en) 1990-06-11 1995-06-07 High affinity PDGF nucleic acid ligands
US08/479,724 Continuation-In-Part US5780228A (en) 1990-06-11 1995-06-07 High affinity nucleic acid ligands to lectins
US08/479,729 Continuation-In-Part US5853984A (en) 1990-06-11 1995-06-07 Use of nucleic acid ligands in flow cytometry
US08/487,426 Continuation-In-Part US5763173A (en) 1990-06-11 1995-06-07 Nucleic acid ligand inhibitors to DNA polymerases
US08/487,425 Continuation-In-Part US5789163A (en) 1990-06-11 1995-06-07 Enzyme linked oligonucleotide assays (ELONAS)
US08/484,552 Continuation-In-Part US5849890A (en) 1990-06-11 1995-06-07 High affinity oligonucleotide ligands to chorionic gonadotropin hormone and related glycoprotein hormones
US08/976,413 Continuation-In-Part US6127119A (en) 1990-06-11 1997-11-21 Nucleic acid ligands of tissue target
US09/860,474 Continuation-In-Part US6696252B2 (en) 1990-06-11 2001-05-18 High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)

Publications (1)

Publication Number Publication Date
US5496938A true US5496938A (en) 1996-03-05

Family

ID=46202104

Family Applications (6)

Application Number Title Priority Date Filing Date
US07/964,624 Expired - Lifetime US5496938A (en) 1990-06-11 1992-10-21 Nucleic acid ligands to HIV-RT and HIV-1 rev
US08/442,062 Expired - Lifetime US5595877A (en) 1990-06-11 1995-05-16 Methods of producing nucleic acid ligands
US08/748,697 Expired - Lifetime US5817785A (en) 1990-06-11 1996-11-13 Methods of producing nucleic acid ligands
US09/165,616 Expired - Fee Related US6344318B1 (en) 1990-06-11 1998-10-02 Methods of producing nucleic acid ligands
US10/040,497 Expired - Fee Related US6716583B2 (en) 1990-06-11 2002-01-07 Methods of producing nucleic acid ligands
US10/818,954 Expired - Fee Related US7368236B2 (en) 1990-06-11 2004-04-06 Methods of producing nucleic acid ligands

Family Applications After (5)

Application Number Title Priority Date Filing Date
US08/442,062 Expired - Lifetime US5595877A (en) 1990-06-11 1995-05-16 Methods of producing nucleic acid ligands
US08/748,697 Expired - Lifetime US5817785A (en) 1990-06-11 1996-11-13 Methods of producing nucleic acid ligands
US09/165,616 Expired - Fee Related US6344318B1 (en) 1990-06-11 1998-10-02 Methods of producing nucleic acid ligands
US10/040,497 Expired - Fee Related US6716583B2 (en) 1990-06-11 2002-01-07 Methods of producing nucleic acid ligands
US10/818,954 Expired - Fee Related US7368236B2 (en) 1990-06-11 2004-04-06 Methods of producing nucleic acid ligands

Country Status (1)

Country Link
US (6) US5496938A (en)

Cited By (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996036692A1 (en) * 1995-05-17 1996-11-21 The Regents Of The University Of California In vivo selection of rna-binding peptides
WO1997009053A1 (en) * 1995-09-08 1997-03-13 Scriptgen Pharmaceuticals, Inc. 2-deoxystreptamine as a molecular scaffold for preparing functionally and spatially diverse molecules and pharmaceutical compositions thereof
WO1997009342A1 (en) * 1995-09-08 1997-03-13 Scriptgen Pharmaceuticals, Inc. Screen for compounds with affinity for rna
WO1997020072A1 (en) * 1995-12-01 1997-06-05 The Penn State Research Foundation Compositions and methods of developing oligonucleotides and oligonucleotide analogs having antiviral activity
US5693502A (en) * 1990-06-11 1997-12-02 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5726017A (en) * 1990-06-11 1998-03-10 Nexstar Pharmaceuticals, Inc. High affinity HIV-1 gag nucleic acid ligands
US5756287A (en) * 1990-06-11 1998-05-26 Nexstar Pharmaceuticals, Inc. High affinity HIV integrase inhibitors
US5763173A (en) * 1990-06-11 1998-06-09 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5766853A (en) * 1990-06-11 1998-06-16 Nexstar Pharmaceuticals, Inc. Method for identification of high affinity nucleic acid ligands to selectins
US5780228A (en) * 1990-06-11 1998-07-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US5786462A (en) * 1990-06-11 1998-07-28 Nexstar Pharmaceuticals, Inc. High affinity ssDNA ligands of HIV-1 reverse transcriptase
US5861254A (en) * 1997-01-31 1999-01-19 Nexstar Pharmaceuticals, Inc. Flow cell SELEX
US5869641A (en) * 1990-06-11 1999-02-09 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of CD4
US5874557A (en) * 1990-06-11 1999-02-23 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US6001988A (en) * 1990-06-11 1999-12-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US6001570A (en) * 1997-02-18 1999-12-14 Invitro Diagnostics, Inc. Compositions, methods, kits and apparatus for determining the presence or absence of target molecules
US6011020A (en) * 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US6020130A (en) * 1995-06-07 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands that bind to and inhibit DNA polymerases
US6140490A (en) * 1996-02-01 2000-10-31 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of complement system proteins
US6177557B1 (en) 1990-06-11 2001-01-23 Nexstar Pharmaceuticals, Inc. High affinity ligands of basic fibroblast growth factor and thrombin
US6183967B1 (en) 1995-06-07 2001-02-06 Nexstar Pharmaceuticals Nucleic acid ligand inhibitors to DNA polymerases
WO2001049244A2 (en) * 2000-01-03 2001-07-12 Yeda Research And Development Co. Ltd. Database system and method useful for predicting putative ligand binding sites
US6261774B1 (en) * 1990-06-11 2001-07-17 Gilead Sciences, Inc. Truncation selex method
US6280932B1 (en) 1990-06-11 2001-08-28 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US6308145B1 (en) * 1992-03-27 2001-10-23 Akiko Itai Methods for searching stable docking models of biopolymer-ligand molecule complex
US6331392B1 (en) 1997-03-05 2001-12-18 Anadys Pharmaceutical, Inc. Screen employing fluorescence anisotropy to identify compounds with affinity for nucleic acids
US6376190B1 (en) 2000-09-22 2002-04-23 Somalogic, Inc. Modified SELEX processes without purified protein
US6376178B1 (en) 1993-09-03 2002-04-23 Duke University Method of nucleic acid sequencing
EP1206575A1 (en) * 1999-07-28 2002-05-22 Gilead Sciences, Inc. Transcription-free selex
US6395888B1 (en) 1996-02-01 2002-05-28 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US20020077306A1 (en) * 1994-07-14 2002-06-20 Ludger Dinkelborg Conjugates made of metal complexes and oligonucleotides, agents containing the conjugates, their use in radiodiagnosis as well as process for their production
US6465188B1 (en) 1990-06-11 2002-10-15 Gilead Sciences, Inc. Nucleic acid ligand complexes
US20030143612A1 (en) * 2001-07-18 2003-07-31 Pointilliste, Inc. Collections of binding proteins and tags and uses thereof for nested sorting and high throughput screening
US20040022727A1 (en) * 2002-06-18 2004-02-05 Martin Stanton Aptamer-toxin molecules and methods for using same
WO2004011680A1 (en) 2002-07-25 2004-02-05 Archemix Corp. Regulated aptamer therapeutics
US20040058884A1 (en) * 1990-06-11 2004-03-25 Brian Hicke Tenasin-C nucleic acid ligands
US20040106145A1 (en) * 1999-01-19 2004-06-03 Somalogic, Inc. Method and apparatus for the automated generation of nucleic acid ligands
US20040137010A1 (en) * 2002-09-17 2004-07-15 Charles Wilson Prophylactic and therapeutic HIV aptamers
US20040180360A1 (en) * 2002-11-21 2004-09-16 Charles Wilson Multivalent aptamer therapeutics with improved pharmacodynamic properties and methods of making and using the same
US20040197804A1 (en) * 2002-12-03 2004-10-07 Keefe Anthony D. Method for in vitro selection of 2'-substituted nucleic acids
US20040247680A1 (en) * 2003-06-06 2004-12-09 Farokhzad Omid C. Targeted delivery of controlled release polymer systems
US20040249130A1 (en) * 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US20040253679A1 (en) * 2002-11-21 2004-12-16 David Epstein Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20040253243A1 (en) * 2003-01-21 2004-12-16 David Epstein Aptamer therapeutics useful in ocular pharmacotherapy
US20050037394A1 (en) * 2002-12-03 2005-02-17 Keefe Anthony D. Method for in vitro selection of 2'-substituted nucleic acids
US20050043265A1 (en) * 1990-06-11 2005-02-24 Gilead Sciences Inc. High affinity RNA ligands of basic fibroblast growth factor
US20050096257A1 (en) * 2003-08-27 2005-05-05 David Shima Combination therapy for the treatment of ocular neovascular disorders
US20050124565A1 (en) * 2002-11-21 2005-06-09 Diener John L. Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20050159351A1 (en) * 2002-11-21 2005-07-21 Dilara Grate Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US6927024B2 (en) 1998-11-30 2005-08-09 Genentech, Inc. PCR assay
US20050250106A1 (en) * 2003-04-24 2005-11-10 David Epstein Gene knock-down by intracellular expression of aptamers
US20050282906A1 (en) * 2004-03-25 2005-12-22 North Shore-Long Island Jewish Research Institute Neural tourniquet
US20060018871A1 (en) * 2004-02-12 2006-01-26 Claude Benedict Aptamer therapeutics useful in the treatment of complement-related disorders
US20060029933A1 (en) * 2004-08-03 2006-02-09 Borer Philip N Branched and multi-chain nucleic acid switches for sensing and screening
US7005260B1 (en) 2000-01-28 2006-02-28 Gilead Sciences, Inc. Tenascin-C nucleic acid ligands
US20060183702A1 (en) * 2004-09-07 2006-08-17 Diener John L Aptamers to von Willebrand factor and their use as thrombotic disease therapeutics
US20060193821A1 (en) * 2004-03-05 2006-08-31 Diener John L Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US20060216692A1 (en) * 2003-02-04 2006-09-28 Borer Philip N Switchable nucleic acids for diagnostics, screening and molecular electronics
WO2006102540A2 (en) * 2005-03-24 2006-09-28 Syracuse University Elucidation of high affinity, high specificity oligonucleotides and derivatized oligonucleotide sequences for target recognition
US20060264369A1 (en) * 2004-09-07 2006-11-23 Diener John L Aptamers to von Willebrand Factor and their use as thrombotic disease therapeutics
US20070009907A1 (en) * 2004-04-26 2007-01-11 Paula Burmeister Nucleic acid ligands specific to immunoglobulin E and their use as atopic disease therapeutics
US20070048248A1 (en) * 2004-02-12 2007-03-01 Claude Benedict Aptamer therapeutics useful in the treatment of complement-related disorders
US20070066550A1 (en) * 2004-03-05 2007-03-22 Diener John L Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US20080140138A1 (en) * 2002-02-26 2008-06-12 Ivanova Svetlana M Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
US20080214489A1 (en) * 2004-04-19 2008-09-04 Anthony Dominic Keefe Aptamer-mediated intracellular delivery of oligonucleotides
EP1975172A2 (en) 1999-07-29 2008-10-01 Gilead Sciences, Inc. High affinity TGFß nucleic acid ligands and inhibitors
US20080249439A1 (en) * 2004-03-25 2008-10-09 The Feinstein Institute For Medical Research Treatment of inflammation by non-invasive stimulation
EP2000534A2 (en) 1995-05-03 2008-12-10 Gilead Sciences, Inc. Nucleic acid ligands of tissue target related applications
EP2011503A2 (en) 1995-06-07 2009-01-07 Gilead Sciences, Inc. High affinity nucleic acid ligands of cytokines
US20090030925A1 (en) * 2007-07-23 2009-01-29 Microsoft Corporation Clustering phylogenetic variation patterns
US20090075342A1 (en) * 2005-04-26 2009-03-19 Sharon Cload Metabolic profile directed aptamer medicinal chemistry
US20090105172A1 (en) * 2005-03-07 2009-04-23 Diener John L Stabilized Aptamers to PSMA and Their Use as Prostate Cancer Therapeutics
US20090118481A1 (en) * 1995-06-07 2009-05-07 Gilead Sciences, Inc. High Affinity Nucleic Acid Ligands To Lectins
EP2070939A1 (en) 2001-05-25 2009-06-17 Duke University Modulators of pharmacological agents
US20090163437A1 (en) * 2007-10-16 2009-06-25 Regado Biosciences, Inc. Steady-state subcutaneous administration of aptamers
US20090203766A1 (en) * 2007-06-01 2009-08-13 Archemix Corp. vWF aptamer formulations and methods for use
US20090247934A1 (en) * 2008-03-31 2009-10-01 Tracey Kevin J Methods and systems for reducing inflammation by neuromodulation of t-cell activity
US20090248097A1 (en) * 2000-05-23 2009-10-01 Feinstein Institute For Medical Research, The Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US20090269356A1 (en) * 2006-03-08 2009-10-29 David Epstein Complement Binding Aptamers and Anti-C5 Agents Useful in the Treatment of Ocular Disorders
US20090275997A1 (en) * 2008-05-01 2009-11-05 Michael Allen Faltys Vagus nerve stimulation electrodes and methods of use
EP2172566A1 (en) 2007-07-17 2010-04-07 Somalogic, Inc. Method for generating aptameters with improved off-rates
US20100125304A1 (en) * 2008-11-18 2010-05-20 Faltys Michael A Devices and methods for optimizing electrode placement for anti-inflamatory stimulation
US7767803B2 (en) 2002-06-18 2010-08-03 Archemix Corp. Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
WO2010096388A2 (en) 2009-02-18 2010-08-26 Carnegie Mellon University Quenched dendrimeric dyes for bright detection
US20100291562A1 (en) * 2007-04-04 2010-11-18 Michael Adler Method for the detection of an analyte in biological matrix
US20100312320A1 (en) * 2009-06-09 2010-12-09 Faltys Michael A Nerve cuff with pocket for leadless stimulator
WO2010141771A1 (en) 2009-06-03 2010-12-09 Regado Biosciences, Inc. Nucleic acid modulators of glycoprotein vi
WO2011006075A1 (en) 2009-07-09 2011-01-13 Somalogic, Inc. Method for generating aptamers with improved off-rates
EP2280083A1 (en) 1997-12-15 2011-02-02 Somalogic, Inc. Nucleid acid ligand diagnostic biochip
US20110104667A1 (en) * 2009-11-03 2011-05-05 Infoscitex Corporation Methods for identifying nucleic acid ligands
US20110104668A1 (en) * 2009-11-03 2011-05-05 Infoscitex Corporation Nucleic acid ligands against infectious prions
US7947447B2 (en) 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
US20110165037A1 (en) * 2010-01-07 2011-07-07 Ismagilov Rustem F Interfaces that eliminate non-specific adsorption, and introduce specific interactions
US20110190849A1 (en) * 2009-12-23 2011-08-04 Faltys Michael A Neural stimulation devices and systems for treatment of chronic inflammation
US7998940B2 (en) 2004-09-07 2011-08-16 Archemix Corp. Aptamers to von Willebrand factor and their use as thrombotic disease therapeutics
US20110212030A1 (en) * 1995-05-04 2011-09-01 Gilead Sciences, Inc. Nucleic Acid Ligand Complexes
WO2011150079A1 (en) 2010-05-25 2011-12-01 Carnegie Mellon University Targeted probes of cellular physiology
US8101385B2 (en) 2005-06-30 2012-01-24 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
US8105813B2 (en) 2005-06-30 2012-01-31 Archemix Corp. Materials and methods for the generation of fully 2′-modified nucleic acid transcripts
EP2436391A2 (en) 2004-11-02 2012-04-04 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US8391970B2 (en) 2007-08-27 2013-03-05 The Feinstein Institute For Medical Research Devices and methods for inhibiting granulocyte activation by neural stimulation
WO2013034687A1 (en) 2011-09-09 2013-03-14 Universität Bern Verwaltungsdirektion Avibacterium paragallinarum rtx toxin
EP2623601A2 (en) 2003-04-21 2013-08-07 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
EP2629094A1 (en) 2007-01-24 2013-08-21 Carnegie Mellon University Optical biosensors
US8598140B2 (en) 2010-04-12 2013-12-03 Somalogic, Inc. Aptamers to β-NGF and their use in treating β-NGF mediated diseases and disorders
WO2014068408A2 (en) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamers and uses thereof
WO2014100434A1 (en) 2012-12-19 2014-06-26 Caris Science, Inc. Compositions and methods for aptamer screening
US8788034B2 (en) 2011-05-09 2014-07-22 Setpoint Medical Corporation Single-pulse activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
WO2015031694A2 (en) 2013-08-28 2015-03-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
US8996116B2 (en) 2009-10-30 2015-03-31 Setpoint Medical Corporation Modulation of the cholinergic anti-inflammatory pathway to treat pain or addiction
WO2015075622A1 (en) 2013-11-21 2015-05-28 Inis Biotech Llc Aptamers against the myelin basic protein as neuroprotective agents
US9211410B2 (en) 2009-05-01 2015-12-15 Setpoint Medical Corporation Extremely low duty-cycle activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US9365855B2 (en) 2010-03-03 2016-06-14 Somalogic, Inc. Aptamers to 4-1BB and their use in treating diseases and disorders
WO2016145128A1 (en) 2015-03-09 2016-09-15 Caris Science, Inc. Oligonucleotide probes and uses thereof
WO2017004243A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
US9572983B2 (en) 2012-03-26 2017-02-21 Setpoint Medical Corporation Devices and methods for modulation of bone erosion
US9662490B2 (en) 2008-03-31 2017-05-30 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation and administration of an anti-inflammatory drug
WO2017181110A1 (en) 2016-04-14 2017-10-19 Fred Hutchinson Cancer Research Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
US9833621B2 (en) 2011-09-23 2017-12-05 Setpoint Medical Corporation Modulation of sirtuins by vagus nerve stimulation
US9910034B2 (en) 2007-11-06 2018-03-06 Ambergen, Inc. Methods and compositions for phototransfer
EP3335714A1 (en) 2011-12-30 2018-06-20 Quest Diagnostics Investments Incorporated Aptamers and diagnostic methods for detecting the egf receptor
US10314501B2 (en) 2016-01-20 2019-06-11 Setpoint Medical Corporation Implantable microstimulators and inductive charging systems
US10329617B2 (en) 2008-05-08 2019-06-25 The Johns Hopkins University Compositions and methods for modulating an immune response
US10434177B2 (en) 2014-11-17 2019-10-08 Carnegie Mellon University Activatable two-component photosensitizers
US10583304B2 (en) 2016-01-25 2020-03-10 Setpoint Medical Corporation Implantable neurostimulator having power control and thermal regulation and methods of use
US10596367B2 (en) 2016-01-13 2020-03-24 Setpoint Medical Corporation Systems and methods for establishing a nerve block
WO2020117914A1 (en) 2018-12-04 2020-06-11 Roche Sequencing Solutions, Inc. Spatially oriented quantum barcoding of cellular targets
US10695569B2 (en) 2016-01-20 2020-06-30 Setpoint Medical Corporation Control of vagal stimulation
US10731166B2 (en) 2016-03-18 2020-08-04 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10912712B2 (en) 2004-03-25 2021-02-09 The Feinstein Institutes For Medical Research Treatment of bleeding by non-invasive stimulation
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
US10945954B2 (en) 2012-02-06 2021-03-16 President And Fellows Of Harvard College ARRDC1-mediated microvesicles (ARMMS) and uses thereof
US11001817B2 (en) 2014-10-31 2021-05-11 President And Fellows Of Harvard College Delivery of cargo proteins via ARRDC1-mediated microvesicles (ARMMs)
US11051744B2 (en) 2009-11-17 2021-07-06 Setpoint Medical Corporation Closed-loop vagus nerve stimulation
US11124822B2 (en) 2014-10-17 2021-09-21 Carnegie Mellon University Enhanced biomolecule detection assays based on tyramide signal amplification and gammaPNA probes
US11173307B2 (en) 2017-08-14 2021-11-16 Setpoint Medical Corporation Vagus nerve stimulation pre-screening test
US11207518B2 (en) 2004-12-27 2021-12-28 The Feinstein Institutes For Medical Research Treating inflammatory disorders by stimulation of the cholinergic anti-inflammatory pathway
US11260229B2 (en) 2018-09-25 2022-03-01 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via coordinated trigeminal and vagal nerve stimulation
US11273171B2 (en) 2013-07-12 2022-03-15 Iveric Bio, Inc. Methods for treating or preventing ophthalmological conditions
US11311725B2 (en) 2014-10-24 2022-04-26 Setpoint Medical Corporation Systems and methods for stimulating and/or monitoring loci in the brain to treat inflammation and to enhance vagus nerve stimulation
US11344724B2 (en) 2004-12-27 2022-05-31 The Feinstein Institutes For Medical Research Treating inflammatory disorders by electrical vagus nerve stimulation
US11406833B2 (en) 2015-02-03 2022-08-09 Setpoint Medical Corporation Apparatus and method for reminding, prompting, or alerting a patient with an implanted stimulator
US11471681B2 (en) 2016-01-20 2022-10-18 Setpoint Medical Corporation Batteryless implantable microstimulators
US11660443B2 (en) 2018-04-20 2023-05-30 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via electrical trigeminal nerve stimulation
EP4219516A2 (en) 2012-07-13 2023-08-02 Wave Life Sciences Ltd. Chiral control
US11730823B2 (en) 2016-10-03 2023-08-22 President And Fellows Of Harvard College Delivery of therapeutic RNAs via ARRDC1-mediated microvesicles
US11938324B2 (en) 2020-05-21 2024-03-26 The Feinstein Institutes For Medical Research Systems and methods for vagus nerve stimulation
US11969253B2 (en) 2021-06-02 2024-04-30 Setpoint Medical Corporation Closed-loop vagus nerve stimulation

Families Citing this family (198)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5496938A (en) 1990-06-11 1996-03-05 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to HIV-RT and HIV-1 rev
US6168778B1 (en) 1990-06-11 2001-01-02 Nexstar Pharmaceuticals, Inc. Vascular endothelial growth factor (VEGF) Nucleic Acid Ligand Complexes
US20060084797A1 (en) * 1990-06-11 2006-04-20 Gilead Sciences, Inc. High affinity TGFbeta nucleic acid ligands and inhibitors
US6147204A (en) * 1990-06-11 2000-11-14 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
JPH0793370A (en) * 1993-09-27 1995-04-07 Hitachi Device Eng Co Ltd Gene data base retrieval system
US5859228A (en) * 1995-05-04 1999-01-12 Nexstar Pharmaceuticals, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
US6699843B2 (en) * 1995-06-07 2004-03-02 Gilead Sciences, Inc. Method for treatment of tumors using nucleic acid ligands to PDGF
US6229002B1 (en) * 1995-06-07 2001-05-08 Nexstar Pharmaceuticlas, Inc. Platelet derived growth factor (PDGF) nucleic acid ligand complexes
US6426335B1 (en) * 1997-10-17 2002-07-30 Gilead Sciences, Inc. Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
US6051698A (en) * 1997-06-06 2000-04-18 Janjic; Nebojsa Vascular endothelial growth factor (VEGF) nucleic acid ligand complexes
AU5927598A (en) * 1997-01-23 1998-08-18 Immusol Incorporated Gene functional analysis and discovery using randomized or target-specific ribozyme gene vector libraries
US5834199A (en) * 1997-04-29 1998-11-10 North Carolina State University Methods of identifying transition metal complexes that selectively cleave regulatory elements of mRNA and uses thereof
US20060057573A1 (en) * 2002-02-15 2006-03-16 Somalogic, Inc Methods and reagents for detecting target binding by nucleic acid ligands
US20070166741A1 (en) 1998-12-14 2007-07-19 Somalogic, Incorporated Multiplexed analyses of test samples
US7393529B2 (en) * 1998-04-09 2008-07-01 Idexx Laboratories, Inc. Methods and compositions for inhibiting binding of IgE to a high affinity receptor
US6180348B1 (en) 1998-04-20 2001-01-30 Weihua Li Method of isolating target specific oligonucleotide ligands
EP2360271A1 (en) 1998-06-24 2011-08-24 Illumina, Inc. Decoding of array sensors with microspheres
US6100035A (en) * 1998-07-14 2000-08-08 Cistem Molecular Corporation Method of identifying cis acting nucleic acid elements
US20040259079A1 (en) * 1998-07-22 2004-12-23 Immusol, Inc. Substantially complete ribozyme libraries
US7666609B1 (en) 1998-12-01 2010-02-23 Shanghai Cp Guojian Pharmaceutical Co. Ltd. Method and composition for diagnosis of melanocytic lesions
US6429027B1 (en) 1998-12-28 2002-08-06 Illumina, Inc. Composite arrays utilizing microspheres
US6794136B1 (en) 2000-11-20 2004-09-21 Sangamo Biosciences, Inc. Iterative optimization in the design of binding proteins
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US20030207295A1 (en) * 1999-04-20 2003-11-06 Kevin Gunderson Detection of nucleic acid reactions on bead arrays
AU4476900A (en) 1999-04-20 2000-11-02 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US8080380B2 (en) * 1999-05-21 2011-12-20 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
US8481268B2 (en) 1999-05-21 2013-07-09 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
WO2001006249A2 (en) 1999-07-02 2001-01-25 Conceptual Mindworks, Inc. Organic semiconductor recognition complex and system
BR0003768A (en) * 1999-08-26 2001-07-03 Tomas Antonio Santa Coloma Monoclonal and polyclonal oligibodies (synthetic reagents similar to antibodies based on oligonucleotides) and a method of producing oligibodies
US6941317B1 (en) 1999-09-14 2005-09-06 Eragen Biosciences, Inc. Graphical user interface for display and analysis of biological sequence data
US20030091999A1 (en) * 1999-10-01 2003-05-15 Zhongping Yu Compositions and methods for identifying polypeptides and nucleic acid molecules
EP1226149A4 (en) * 1999-10-01 2003-05-14 Zhongping Yu Compositions and methods for identifying polypeptides and nucleic acid molecules
US7645743B2 (en) * 1999-12-22 2010-01-12 Altermune, Llc Chemically programmable immunity
CA2328356A1 (en) * 1999-12-22 2001-06-22 Itty Atcravi Recreational vehicles
EP1967595A3 (en) 2000-02-16 2008-12-03 Illumina, Inc. Parallel genotyping of multiple patient samples
US20050239061A1 (en) * 2000-03-01 2005-10-27 Marshall William S Identification and use of effectors and allosteric molecules for the alteration of gene expression
US7682837B2 (en) * 2000-05-05 2010-03-23 Board Of Trustees Of Leland Stanford Junior University Devices and methods to form a randomly ordered array of magnetic beads and uses thereof
US20030207271A1 (en) * 2000-06-30 2003-11-06 Holwitt Eric A. Methods and compositions for biological sensors
US7892484B2 (en) * 2000-06-30 2011-02-22 Conceptual Mindworks, Inc. Methods and compositions for neutralizing anthrax and other bioagents
EP2351855A1 (en) 2000-09-26 2011-08-03 Duke University RNA aptamers and methods for identifying the same
DE10049074A1 (en) * 2000-10-02 2002-04-18 Andreas Kage Process for the selection of nucleic acids that bind highly affine to a target
DE10048944A1 (en) * 2000-10-03 2002-04-18 Andreas Kage Process for the selection of nucleic acids that bind highly affine to a target by two-dimensional separation
ATE312942T1 (en) * 2000-10-25 2005-12-15 Hoffmann La Roche AMPLIFICATION USING MODIFIED PRIMERS
JP3578098B2 (en) * 2001-03-16 2004-10-20 富士ゼロックス株式会社 Manufacturing method of electrical connector, electrical connector, and electrical wiring method
WO2003029492A1 (en) * 2001-09-28 2003-04-10 Justin Gallivan Metabolic genes and related methods and compositions
US7781396B2 (en) * 2002-01-31 2010-08-24 Tel Aviv University Future Technology Development L.P. Peptides directed for diagnosis and treatment of amyloid-associated disease
US20040052928A1 (en) * 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
WO2003069333A1 (en) * 2002-02-14 2003-08-21 Illumina, Inc. Automated information processing in randomly ordered arrays
AU2003217580A1 (en) * 2002-02-19 2003-09-09 Syntherica Corporation Surrogate antibodies and methods of preparation and use thereof
EP1572719A4 (en) 2002-02-20 2007-11-14 Univ Minnesota Partial peptide mimetics and methods
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US7591994B2 (en) 2002-12-13 2009-09-22 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US7211382B2 (en) * 2002-04-09 2007-05-01 Orchid Cellmark Inc. Primer extension using modified nucleotides
JP2005538380A (en) 2002-09-11 2005-12-15 ユニヴァーシティ オヴ フロリダ Analysis of nerve cell damage
EP1585824A4 (en) 2002-11-21 2008-01-02 Epict Technologies Preparation and use of single-stranded transcription substrates for synthesis of transcription products corresponding to target sequences
US7491699B2 (en) 2002-12-09 2009-02-17 Ramot At Tel Aviv University Ltd. Peptide nanostructures and methods of generating and using the same
WO2004053099A2 (en) * 2002-12-10 2004-06-24 The Scripps Research Institute A method for treatment of drug addiction and for screening of pharmaceutical agents therefor
ATE426575T1 (en) * 2003-01-07 2009-04-15 Univ Ramot PEPTIDE ANOSTRUCTURES CONTAINING FOREIGN MATERIAL AND METHOD FOR PRODUCING THE SAME
US6943768B2 (en) 2003-02-21 2005-09-13 Xtellus Inc. Thermal control system for liquid crystal cell
US7140685B2 (en) * 2003-03-11 2006-11-28 Gardner Kenneth J Swivel chair joint
US20050232926A1 (en) * 2003-06-06 2005-10-20 Oncomax Acquisition Corp. Antibodies specific for cancer associated antigen SM5-1 and uses thereof
CN1279056C (en) * 2003-06-06 2006-10-11 马菁 Specific antibody of tumor-associated antigen SM5-1 and use thereof
CA2535799A1 (en) 2003-08-15 2005-03-03 University Of Florida Identification of porphyromonas gingivalis virulence polynucleotides for diagnosis, treatment, and monitoring of periodontal diseases
CA2540407A1 (en) * 2003-09-25 2005-03-31 Tel Aviv University Future Technology Development L.P. Compositions and methods using same for treating amyloid-associated diseases
US7625707B2 (en) * 2003-10-02 2009-12-01 Ramot At Tel Aviv University Ltd. Antibacterial agents and methods of identifying and utilizing same
WO2005051174A2 (en) * 2003-11-21 2005-06-09 The Trustees Of Columbia University In The City Of New York Nucleic acid aptamer-based compositions and methods
CA2545006C (en) * 2003-12-12 2013-09-17 Saint Louis University Biosensors for detecting macromolecules and other analytes
PT1745062E (en) * 2004-04-22 2014-07-11 Regado Biosciences Inc Improved modulators of coagulation factors
US20090156471A1 (en) * 2004-07-15 2009-06-18 Ramot At Tel Aviv University Ltd. Use of anti-amyloid agents for treating and typing pathogen infections
WO2006013552A2 (en) 2004-08-02 2006-02-09 Ramot At Tel Aviv University Ltd. Articles of peptide nanostructures and method of forming the same
US7732479B2 (en) 2004-08-19 2010-06-08 Tel Aviv University Future Technology Development L.P. Compositions for treating amyloid associated diseases
WO2006027780A2 (en) * 2004-09-08 2006-03-16 Ramot At Tel Aviv University Ltd. Peptide nanostructures containing end-capping modified peptides and methods of generating and using the same
WO2007001448A2 (en) * 2004-11-04 2007-01-04 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
EP1838350B1 (en) 2005-01-20 2014-08-20 Ryboquin Company Limited Modulators of itch ubiquitinase activity
CA2608429A1 (en) 2005-05-11 2006-11-23 Anthony Albino Reduced risk tobacco products and methods of making same
US7795009B2 (en) * 2005-06-15 2010-09-14 Saint Louis University Three-component biosensors for detecting macromolecules and other analytes
US8956857B2 (en) 2005-06-06 2015-02-17 Mediomics, Llc Three-component biosensors for detecting macromolecules and other analytes
CA2611198C (en) 2005-06-10 2015-04-28 Saint Louis University Methods for the selection of aptamers
US7811809B2 (en) * 2005-06-15 2010-10-12 Saint Louis University Molecular biosensors for use in competition assays
US10004828B2 (en) * 2005-10-11 2018-06-26 Romat at Tel-Aviv University Ltd. Self-assembled Fmoc-ff hydrogels
US7879212B2 (en) * 2005-11-03 2011-02-01 Ramot At Tel-Aviv University Ltd. Peptide nanostructure-coated electrodes
US7576037B2 (en) * 2005-11-18 2009-08-18 Mei Technologies, Inc. Process and apparatus for combinatorial synthesis
WO2007070682A2 (en) * 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
EP2674440B1 (en) 2005-12-16 2019-07-03 IBC Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US8460878B2 (en) 2006-02-21 2013-06-11 The Trustees Of Tufts College Methods and arrays for detecting cells and cellular components in small defined volumes
CA2648099C (en) 2006-03-31 2012-05-29 The Brigham And Women's Hospital, Inc System for targeted delivery of therapeutic agents
CA2652280C (en) 2006-05-15 2014-01-28 Massachusetts Institute Of Technology Polymers for functional particles
US20110052697A1 (en) * 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US9102948B2 (en) 2006-11-17 2015-08-11 22Nd Century Limited, Llc Regulating alkaloids
WO2008147456A2 (en) * 2006-11-20 2008-12-04 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
US9938641B2 (en) * 2006-12-18 2018-04-10 Fluidigm Corporation Selection of aptamers based on geometry
US20110136099A1 (en) * 2007-01-16 2011-06-09 Somalogic, Inc. Multiplexed Analyses of Test Samples
US9725713B1 (en) 2007-01-22 2017-08-08 Steven A Benner In vitro selection with expanded genetic alphabets
US8586303B1 (en) 2007-01-22 2013-11-19 Steven Albert Benner In vitro selection with expanded genetic alphabets
EP2134830A2 (en) * 2007-02-09 2009-12-23 Massachusetts Institute of Technology Oscillating cell culture bioreactor
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
WO2008124639A2 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
JP5658031B2 (en) 2007-06-22 2015-01-21 ボード・オブ・リージエンツ,ザ・ユニバーシテイ・オブ・テキサス・システム Formation of stable submicron peptide or protein particles by thin film freezing
US8404830B2 (en) * 2007-07-17 2013-03-26 Somalogic, Inc. Method for generating aptamers with improved off-rates
ES2627292T3 (en) 2007-10-12 2017-07-27 Massachusetts Institute Of Technology Vaccine Nanotechnology
US8518640B2 (en) * 2007-10-29 2013-08-27 Complete Genomics, Inc. Nucleic acid sequencing and process
ES2916498T3 (en) 2007-12-06 2022-07-01 Genalyte Inc Method for identifying a nucleotide sequence in an unknown species of nucleic acid; and device for carrying out the method
US20090233295A1 (en) * 2008-01-29 2009-09-17 Elias Georges Trim59 directed diagnostics for neoplastic disease
EP2245466A2 (en) 2008-02-04 2010-11-03 Banyan Biomarkers, Inc. Process to diagnose or treat brain injury
US20100048913A1 (en) 2008-03-14 2010-02-25 Angela Brodie Novel C-17-Heteroaryl Steroidal CYP17 Inhibitors/Antiandrogens;Synthesis In Vitro Biological Activities, Pharmacokinetics and Antitumor Activity
EP2285953B1 (en) 2008-05-02 2015-01-14 Cellscript, Llc Rna polyphosphatase compositions, kits, and uses thereof
ES2434742T3 (en) 2008-05-08 2013-12-17 Alellyx Applied Genomics Genes and procedures to increase disease resistance in plants
WO2010005567A2 (en) 2008-07-08 2010-01-14 Oncomed Pharmaceuticals, Inc. Notch1 receptor binding agents and methods of use thereof
AU2009270682B2 (en) 2008-07-17 2016-04-21 Ikfe Lnstitut Fur Klinische Forschung Und Entwicklung Gmbh Biomarkers for insulin resistance and beta-cell dysfunction
EP2338051B1 (en) 2008-07-17 2013-11-20 IKFE Institut für klinische Forschung und Entwicklung GmbH Biomarkers for cardiodiabetes
ES2665245T3 (en) 2008-08-11 2018-04-25 Banyan Biomarkers, Inc. Biomarker detection process and neurological status test
WO2010042933A2 (en) 2008-10-10 2010-04-15 Northwestern University Inhibition and treatment of prostate cancer metastasis
US8277812B2 (en) * 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
EP2347247B1 (en) 2008-10-27 2019-06-26 Genalyte, Inc. Biosensors based on optical probing and sensing
WO2010059820A1 (en) 2008-11-21 2010-05-27 Saint Louis University Biosensor for detecting multiple epitopes on a target
CA2782776A1 (en) 2008-12-04 2010-06-10 Ikfe Gmbh Biomarkers for atherosclerosis
US20110118134A1 (en) 2008-12-11 2011-05-19 Ikfe Gmbh Biomarkers for insulin sensitizer drug response
US20100209350A1 (en) 2008-12-30 2010-08-19 Andreas Pfuetzner Biomarkers for Adipose Tissue Activity
WO2010079428A2 (en) 2009-01-07 2010-07-15 Ikfe Gmbh Biomarkers for appetite regulation
BRPI1008745A2 (en) 2009-02-05 2019-09-17 Tokai Pharmaceuticals Inc cyp17 / antiandrogen steroid inhibitor prodrugs
DK3912643T3 (en) 2009-02-13 2022-10-17 Immunomedics Inc IMMUNOCONJUGATES WITH AN INTRACELLULAR-CLEAVABLE BOND
BRPI1011836A2 (en) * 2009-04-21 2017-05-16 Selecta Biosciences Inc immunotherapeutic agents that provide a th1-induced response
BRPI1011269A2 (en) * 2009-05-05 2016-09-27 Altermune Technologies Llc chemically programmable immunity
KR20120016297A (en) 2009-06-01 2012-02-23 코넬 유니버시티 Integrated optofluidic system using microspheres
WO2010141928A2 (en) 2009-06-05 2010-12-09 University Of Florida Research Foundation, Inc. Isolation and targeted suppression of lignin biosynthetic genes from sugarcane
CA2766057A1 (en) 2009-06-19 2010-12-23 Banyan Biomarkers, Inc. Biomarker assay of neurological condition
US9523701B2 (en) 2009-07-29 2016-12-20 Dynex Technologies, Inc. Sample plate systems and methods
GB0913258D0 (en) 2009-07-29 2009-09-02 Dynex Technologies Inc Reagent dispenser
US9250234B2 (en) 2011-01-19 2016-02-02 Ohmx Corporation Enzyme triggered redox altering chemical elimination (E-TRACE) immunoassay
JP5352742B2 (en) * 2009-08-07 2013-11-27 オームクス コーポレーション Enzymatic redox-change chemical elimination (E-TRACE) immunoassay
HUE040281T2 (en) 2009-09-14 2019-03-28 Banyan Biomarkers Inc Autoantibody markers for diagnosis of traumatic brain injury
US9134323B2 (en) 2009-12-01 2015-09-15 The Rockefeller University Antibody that specifically binds to an epitope in the tureet region of a human Kir channel
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
EP2529033B1 (en) 2010-01-26 2017-05-24 National Jewish Health Methods for risk prediction, diagnosis, prognosis of pulmonary disorders
WO2011100561A1 (en) 2010-02-12 2011-08-18 Saint Louis University Molecular biosensors capable of signal amplification
KR20230044026A (en) 2010-02-24 2023-03-31 이뮤노젠 아이엔씨 Folate receptor 1 antibodies and immunoconjugates and uses thereof
US9678068B2 (en) 2010-03-01 2017-06-13 Quanterix Corporation Ultra-sensitive detection of molecules using dual detection methods
ES2544635T3 (en) 2010-03-01 2015-09-02 Quanterix Corporation Methods to extend the dynamic range in assays for the detection of molecules or particles
US8236574B2 (en) 2010-03-01 2012-08-07 Quanterix Corporation Ultra-sensitive detection of molecules or particles using beads or other capture objects
BR112012023010A8 (en) 2010-03-12 2017-12-26 Immunogen Inc ANTIBODY OR ANTIGEN-BINDING FRAGMENT OF THE SAME THAT SPECIFICALLY BINDS TO CD37, IMMUNOCONJUGATE COMPRISING THE SAME, USE OF THE SAID ANTIBODY, FRAGMENT AND IMMUNOCONJUGATE, COMPOSITION COMPRISING THE SAME, KIT, ISOLATED CELL, AS WELL AS IN VITRO METHOD TO INHIBIT GROWTH MENT OF A CELL THAT EXPRESSES CD37
US20130029362A1 (en) 2010-04-01 2013-01-31 Andreas Jeromin Markers and assays for detection of neurotoxicity
WO2012019024A2 (en) 2010-08-04 2012-02-09 Immunogen, Inc. Her3-binding molecules and immunoconjugates thereof
EP2603607B1 (en) 2010-08-11 2016-04-06 Celula, Inc. Genotyping dna
WO2012051519A2 (en) 2010-10-14 2012-04-19 The Johns Hopkins University Biomarkers of brain injury
CA3060724A1 (en) 2010-11-05 2012-05-10 Genalyte, Inc. Optical analyte detection systems and methods of use
WO2012155134A2 (en) 2011-05-12 2012-11-15 The Johns Hopkins University Assay reagents for a neurogranin diagnostic kit
GB201114662D0 (en) 2011-08-24 2011-10-12 Altermune Technologies Llc Chemically programmable immunity
EP2768967B1 (en) 2011-10-17 2017-12-06 Ohmx Corporation Single, direct detection of hemoglobin a1c percentage using enzyme triggered redox altering chemical elimination (e-trace) immunoassay
US9340567B2 (en) 2011-11-04 2016-05-17 Ohmx Corporation Chemistry used in biosensors
JP6276706B2 (en) 2012-01-09 2018-02-07 オームクス コーポレイション Enzyme cascade method for E-TRACE assay signal amplification
EP2825885B1 (en) 2012-03-12 2021-05-12 The Board of Trustees of the University of Illinois Optical analyte detection systems with magnetic enhancement
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
US9404883B2 (en) 2012-07-27 2016-08-02 Ohmx Corporation Electronic measurements of monolayers following homogeneous reactions of their components
JP2015529809A (en) 2012-07-27 2015-10-08 オームクス コーポレイション Electrical measurement of monolayers after pre-cleavage detection of the presence and activity of enzymes and other target analytes
CA2880833A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
EP2912432B1 (en) 2012-10-24 2018-07-04 Genmark Diagnostics Inc. Integrated multiplex target analysis
PT2900277T (en) 2012-12-13 2022-05-25 Immunomedics Inc Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
EP2774930A1 (en) 2013-03-07 2014-09-10 Aptenia S.R.L. Metallocene compounds and labeled molecules comprising the same for in vivo imaging.
SG11201507093WA (en) 2013-03-14 2015-10-29 Univ Maryland Baltimore Office Of Technology Transfer Androgen receptor down-regulating agents and uses thereof
WO2014143637A1 (en) 2013-03-15 2014-09-18 The Board Of Trustees Of The University Of Illinois Methods and compositions for enhancing immunoassays
RU2015144149A (en) 2013-03-15 2017-04-21 Интермьюн, Инк. PROTEOMIC MARKERS ILF
EP3757226A3 (en) 2013-07-17 2021-05-05 The Johns Hopkins University A multi-protein biomarker assay for brain injury detection and outcome
CN105636594A (en) 2013-08-12 2016-06-01 托凯药业股份有限公司 Biomarkers for treatment of neoplastic disorders using androgen-targeted therapies
EP3071588A4 (en) 2013-11-21 2017-08-02 Somalogic, Inc. Cytidine-5-carboxamide modified nucleotide compositions and methods related thereto
CN105899537A (en) 2014-01-10 2016-08-24 博笛生物科技(北京)有限公司 Compounds and compositions for treating EGFR expressing tumors
US10619210B2 (en) 2014-02-07 2020-04-14 The Johns Hopkins University Predicting response to epigenetic drug therapy
US9880121B2 (en) 2014-04-29 2018-01-30 Ohmx Corporation Bioelectronic binding assay using peak profiling
CN112546231A (en) 2014-07-09 2021-03-26 博笛生物科技有限公司 Combination therapeutic compositions and combination therapeutic methods for treating cancer
CN105440135A (en) 2014-09-01 2016-03-30 博笛生物科技有限公司 Anti-PD-L1 conjugate for treating tumors
CA2954446A1 (en) 2014-07-09 2016-01-14 Shanghai Birdie Biotech, Inc. Anti-pd-l1 combinations for treating tumors
US20160041118A1 (en) 2014-08-11 2016-02-11 Ohmx Corporation Enzyme triggered redox altering chemical elimination (e-trace) assay with multiplexing capabilities
EP3191843A1 (en) 2014-09-12 2017-07-19 Mediomics, LLC Molecular biosensors with a modular design
WO2016044697A1 (en) 2014-09-19 2016-03-24 The Johns Hopkins University Biomarkers of cognitive dysfunction
WO2016134365A1 (en) 2015-02-20 2016-08-25 The Johns Hopkins University Biomarkers of myocardial injury
AU2016243036B2 (en) 2015-04-03 2022-02-17 Abbott Laboratories Devices and methods for sample analysis
CA2981512A1 (en) 2015-04-03 2016-10-06 Abbott Laboratories Devices and methods for sample analysis
CA2985818A1 (en) 2015-05-31 2016-12-08 Curegenix Corporation Combination compositions comprising an antagonist of porcupine and a pd-l/pd-1 axis antagonist for immunotherapy
PT3313443T (en) 2015-06-25 2023-08-30 Immunomedics Inc Combining anti-hla-dr or anti-trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
WO2017004463A1 (en) 2015-07-01 2017-01-05 Abbott Laboratories Devices and methods for sample analysis
WO2017187426A1 (en) 2016-04-29 2017-11-02 Aptamir Therapeutics, Inc. Inhibition of mir-22 mirna by apt-110
US11268135B2 (en) 2016-05-10 2022-03-08 Pattern Bioscience, Inc. Compositions and methods for identifying, quantifying, and/or characterizing an analyte
US11041863B2 (en) 2016-07-21 2021-06-22 Retinal Solutions Llc Frizzled-4 mutation indicator of retinopathy and intrauterine growth restriction
WO2018067878A1 (en) 2016-10-05 2018-04-12 Abbott Laboratories Devices and methods for sample analysis
AU2017338914B2 (en) 2016-10-05 2023-07-13 Abbott Laboratories Devices and methods for sample analysis
US20200171024A1 (en) 2017-06-26 2020-06-04 The Regents Of The University Of Colorado, A Body Corporate Biomarkers for the diagnosis and treatment of fibrotic lung disease
US11725056B2 (en) 2017-10-03 2023-08-15 Cedars-Sinai Medical Center Methods for targeting the immune checkpoint PD1 pathway for treating pulmonary fibrosis
BR112020024715A2 (en) 2018-06-04 2021-03-23 Avon Products. Inc. protein biomarkers to identify and treat skin aging and skin conditions
JP7379505B2 (en) 2018-10-02 2023-11-14 ウェアオプティモ ピーティーワイ リミテッド Determining fluid level
WO2020144535A1 (en) 2019-01-08 2020-07-16 Aduro Biotech Holdings, Europe B.V. Methods and compositions for treatment of multiple myeloma
US20220276256A1 (en) 2019-08-07 2022-09-01 Abbott Laboratories Methods for detecting assay interferents and increasing dynamic range
WO2021026402A1 (en) 2019-08-07 2021-02-11 Abbott Laboratories Chemiluminescent compounds for multiplexing
US20230096625A1 (en) 2020-03-10 2023-03-30 Abbott Laboratories Method for droplet loading into nanowells
GB2594094A (en) 2020-04-17 2021-10-20 Pockit Diagnostics Ltd Method for diagnosing stroke caused by large vessel occlusion
WO2024030432A1 (en) 2022-08-01 2024-02-08 Gensaic, Inc. Therapeutic phage-derived particles

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2183661B (en) * 1985-03-30 1989-06-28 Marc Ballivet Method for obtaining dna, rna, peptides, polypeptides or proteins by means of a dna recombinant technique
US4904582A (en) * 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
WO1989006694A1 (en) * 1988-01-15 1989-07-27 Trustees Of The University Of Pennsylvania Process for selection of proteinaceous substances which mimic growth-inducing molecules
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
EP0533838B1 (en) * 1990-06-11 1997-12-03 NeXstar Pharmaceuticals, Inc. Nucleic acid ligands
US5496938A (en) * 1990-06-11 1996-03-05 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands to HIV-RT and HIV-1 rev
US5270163A (en) * 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
US5527894A (en) * 1990-06-11 1996-06-18 Nexstar Pharmacueticals, Inc. Ligands of HIV-1 tat protein
US5660985A (en) * 1990-06-11 1997-08-26 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands containing modified nucleotides
CA2104698A1 (en) * 1991-02-21 1992-08-22 John J. Toole Aptamers specific for biomolecules and methods of making
JPH07501929A (en) 1991-08-23 1995-03-02 アイシス・ファーマシューティカルス・インコーポレーテッド Synthetic non-randomization of oligomeric fragments
JPH10242773A (en) * 1997-02-27 1998-09-11 Oki Electric Ind Co Ltd Feedback amplifier circuit

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
Bass and Cech (1984) Nature 308:820. *
Carey et al. (1983) Biochemistry 22:2601. *
Joyce (1989) in RNA: Catalysis, Splicing, Evolution, Belfort and Shub (eds.), Elsevier, Amsterdam pp. 83 87. *
Joyce (1989) in RNA: Catalysis, Splicing, Evolution, Belfort and Shub (eds.), Elsevier, Amsterdam pp. 83-87.
Kacian et al. (1972) Proc. Natl. Acad. Sci. USA 69:3038. *
Kinzler et al. "Wholegenome ICR: Application to the Identification of Sequences Bound on Gene Regulatory Proteins", Nuc. Acids Res. 17(10): 3645-3653 (1989).
Kinzler et al. Wholegenome ICR: Application to the Identification of Sequences Bound on Gene Regulatory Proteins , Nuc. Acids Res. 17(10): 3645 3653 (1989). *
Kramer et al. (1974) J. Mol. Biol. 89:719. *
Levisohn and Spiegelman (1968) Proc. Natl. Acad. Sci. USA 60:866. *
Levisohn and Spiegelman (1969) Proc. Natl. Acad. Sci. USA 63:805. *
Mills et al. (1967) Proc. Natl. Acad. Sci. USA 58:217. *
Mills et al. (1973) Science 180:916. *
Rich et al. (1984) Ann. Rev. Biochem. 53:791. *
Robertson and Joyce (1990) Nature 344:467. *
Romaniuk et al. (1987) Biochemistry 26:1563. *
Saffhill et al. (1970) J. Mol. Biol. 51:531. *
Schimmel (1989) Cell 58:9. *
Tuerk et al. (1988) Proc. Natl. Acad. Sci. USA 85:1364. *
Uhlenbeck et al. (1983) J. Biomol. Structure and Dynamics 1:539. *
Witherell and Uhlenbeck (1989) Biochemistry 28:71. *
Yarus (1988) Science 240:1751. *

Cited By (282)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050164974A1 (en) * 1990-06-11 2005-07-28 Larry Gold Nucleic acid ligand complexes
US6855496B2 (en) 1990-06-11 2005-02-15 Gilead Sciences, Inc. Truncation SELEX method
US5869641A (en) * 1990-06-11 1999-02-09 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of CD4
US20030191084A1 (en) * 1990-06-11 2003-10-09 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US5693502A (en) * 1990-06-11 1997-12-02 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5726017A (en) * 1990-06-11 1998-03-10 Nexstar Pharmaceuticals, Inc. High affinity HIV-1 gag nucleic acid ligands
US20030125263A1 (en) * 1990-06-11 2003-07-03 Gilead Sciences, Inc. Nucleic acid ligand complexes
US5763173A (en) * 1990-06-11 1998-06-09 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US5766853A (en) * 1990-06-11 1998-06-16 Nexstar Pharmaceuticals, Inc. Method for identification of high affinity nucleic acid ligands to selectins
US5780228A (en) * 1990-06-11 1998-07-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US5786462A (en) * 1990-06-11 1998-07-28 Nexstar Pharmaceuticals, Inc. High affinity ssDNA ligands of HIV-1 reverse transcriptase
US6544959B1 (en) 1990-06-11 2003-04-08 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US20040043923A1 (en) * 1990-06-11 2004-03-04 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US6465188B1 (en) 1990-06-11 2002-10-15 Gilead Sciences, Inc. Nucleic acid ligand complexes
US5874557A (en) * 1990-06-11 1999-02-23 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand inhibitors to DNA polymerases
US7399752B2 (en) 1990-06-11 2008-07-15 Gilead Science, Inc. High affinity nucleic acid ligands to lectins
US5756287A (en) * 1990-06-11 1998-05-26 Nexstar Pharmaceuticals, Inc. High affinity HIV integrase inhibitors
US6001988A (en) * 1990-06-11 1999-12-14 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands to lectins
US20050136474A1 (en) * 1990-06-11 2005-06-23 Gilead Sciences, Inc. Truncation SELEX method
US6011020A (en) * 1990-06-11 2000-01-04 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US6280932B1 (en) 1990-06-11 2001-08-28 Gilead Sciences, Inc. High affinity nucleic acid ligands to lectins
US20040058884A1 (en) * 1990-06-11 2004-03-25 Brian Hicke Tenasin-C nucleic acid ligands
US7196069B2 (en) 1990-06-11 2007-03-27 Gilead Sciences, Inc. High affinity RNA ligands of basic fibroblast growth factor
US6177557B1 (en) 1990-06-11 2001-01-23 Nexstar Pharmaceuticals, Inc. High affinity ligands of basic fibroblast growth factor and thrombin
US20040072234A1 (en) * 1990-06-11 2004-04-15 Parma David H. High affinity nucleic acid ligands to lectins
US20050043265A1 (en) * 1990-06-11 2005-02-24 Gilead Sciences Inc. High affinity RNA ligands of basic fibroblast growth factor
US6261774B1 (en) * 1990-06-11 2001-07-17 Gilead Sciences, Inc. Truncation selex method
US6308145B1 (en) * 1992-03-27 2001-10-23 Akiko Itai Methods for searching stable docking models of biopolymer-ligand molecule complex
US6808897B2 (en) 1993-09-03 2004-10-26 Duke University Method of nucleic acid sequencing
US6376178B1 (en) 1993-09-03 2002-04-23 Duke University Method of nucleic acid sequencing
US20020077306A1 (en) * 1994-07-14 2002-06-20 Ludger Dinkelborg Conjugates made of metal complexes and oligonucleotides, agents containing the conjugates, their use in radiodiagnosis as well as process for their production
EP2000534A2 (en) 1995-05-03 2008-12-10 Gilead Sciences, Inc. Nucleic acid ligands of tissue target related applications
US20110212030A1 (en) * 1995-05-04 2011-09-01 Gilead Sciences, Inc. Nucleic Acid Ligand Complexes
US8071737B2 (en) 1995-05-04 2011-12-06 Glead Sciences, Inc. Nucleic acid ligand complexes
WO1996036692A1 (en) * 1995-05-17 1996-11-21 The Regents Of The University Of California In vivo selection of rna-binding peptides
US5834184A (en) * 1995-05-17 1998-11-10 Harada; Kazuo In vivo selection of RNA-binding peptides
US6183967B1 (en) 1995-06-07 2001-02-06 Nexstar Pharmaceuticals Nucleic acid ligand inhibitors to DNA polymerases
US6020130A (en) * 1995-06-07 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleic acid ligands that bind to and inhibit DNA polymerases
EP2080813A1 (en) 1995-06-07 2009-07-22 Gilead Sciences, Inc. Nucleic acid ligands that bind to and inhibit DNA polymerases
US20090118481A1 (en) * 1995-06-07 2009-05-07 Gilead Sciences, Inc. High Affinity Nucleic Acid Ligands To Lectins
EP2011503A2 (en) 1995-06-07 2009-01-07 Gilead Sciences, Inc. High affinity nucleic acid ligands of cytokines
US6140361A (en) * 1995-09-08 2000-10-31 Scriptgen Pharmaceuticals, Inc. Use of 2-deoxystreptamine as a molecular scaffold for the preparation of functionally and spatially diverse molecules and pharmaceutical compositions thereof
US5942547A (en) * 1995-09-08 1999-08-24 Scriptgen Pharmaceuticals, Inc. 2-Deoxystreptamine derivatives, pharmaceutical compositions thereof and therapeutic methods using same
WO1997009053A1 (en) * 1995-09-08 1997-03-13 Scriptgen Pharmaceuticals, Inc. 2-deoxystreptamine as a molecular scaffold for preparing functionally and spatially diverse molecules and pharmaceutical compositions thereof
US20030113779A1 (en) * 1995-09-08 2003-06-19 Arenas Jaime E. Screen for compounds with affinity for nucleic acids
US6337183B1 (en) 1995-09-08 2002-01-08 Scriptgen Pharmaceuticals, Inc. Screen for compounds with affinity for nucleic acids
WO1997009342A1 (en) * 1995-09-08 1997-03-13 Scriptgen Pharmaceuticals, Inc. Screen for compounds with affinity for rna
US6503721B2 (en) 1995-09-08 2003-01-07 Anadys Pharmaceuticals, Inc. Screen for compounds with affinity for nucleic acids
US5856085A (en) * 1995-12-01 1999-01-05 The Penn State Research Foundation Compositions and methods of developing oligonucleotides and oligonucleotide analogs having antiviral activity
WO1997020072A1 (en) * 1995-12-01 1997-06-05 The Penn State Research Foundation Compositions and methods of developing oligonucleotides and oligonucleotide analogs having antiviral activity
US20060079477A1 (en) * 1996-02-01 2006-04-13 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US6395888B1 (en) 1996-02-01 2002-05-28 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US6566343B2 (en) 1996-02-01 2003-05-20 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US6140490A (en) * 1996-02-01 2000-10-31 Nexstar Pharmaceuticals, Inc. High affinity nucleic acid ligands of complement system proteins
US7964572B2 (en) 1996-02-01 2011-06-21 Gilead Sciences, Inc. High affinity nucleic acid ligands of complement system proteins
US20110196021A1 (en) * 1996-02-01 2011-08-11 Gilead Sciences, Inc. High Affinity Nucleic Acid Ligands of Complement System Proteins
US5861254A (en) * 1997-01-31 1999-01-19 Nexstar Pharmaceuticals, Inc. Flow cell SELEX
US6001570A (en) * 1997-02-18 1999-12-14 Invitro Diagnostics, Inc. Compositions, methods, kits and apparatus for determining the presence or absence of target molecules
US20030152992A1 (en) * 1997-03-05 2003-08-14 Laing Lance G. Screen employing fluorescence anisotropy to identify compounds with affinity for nucleic acids
US6569628B2 (en) 1997-03-05 2003-05-27 Anadys Pharmaceuticals, Inc. Screen employing fluorescence anisotropy to identify compounds with affinity for nucleic acids
US6331392B1 (en) 1997-03-05 2001-12-18 Anadys Pharmaceutical, Inc. Screen employing fluorescence anisotropy to identify compounds with affinity for nucleic acids
EP2280083A1 (en) 1997-12-15 2011-02-02 Somalogic, Inc. Nucleid acid ligand diagnostic biochip
US6927024B2 (en) 1998-11-30 2005-08-09 Genentech, Inc. PCR assay
US7629151B2 (en) 1999-01-19 2009-12-08 Somalogic, Inc. Method and apparatus for the automated generation of nucleic acid ligands
US20040106145A1 (en) * 1999-01-19 2004-06-03 Somalogic, Inc. Method and apparatus for the automated generation of nucleic acid ligands
EP1206575A4 (en) * 1999-07-28 2004-12-01 Gilead Sciences Inc Transcription-free selex
EP1206575A1 (en) * 1999-07-28 2002-05-22 Gilead Sciences, Inc. Transcription-free selex
EP1975172A2 (en) 1999-07-29 2008-10-01 Gilead Sciences, Inc. High affinity TGFß nucleic acid ligands and inhibitors
WO2001049244A2 (en) * 2000-01-03 2001-07-12 Yeda Research And Development Co. Ltd. Database system and method useful for predicting putative ligand binding sites
WO2001049244A3 (en) * 2000-01-03 2002-03-07 Yeda Res & Dev Database system and method useful for predicting putative ligand binding sites
US7005260B1 (en) 2000-01-28 2006-02-28 Gilead Sciences, Inc. Tenascin-C nucleic acid ligands
US8914114B2 (en) 2000-05-23 2014-12-16 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US10166395B2 (en) 2000-05-23 2019-01-01 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US9987492B2 (en) 2000-05-23 2018-06-05 The Feinstein Institute For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US20090248097A1 (en) * 2000-05-23 2009-10-01 Feinstein Institute For Medical Research, The Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US10561846B2 (en) 2000-05-23 2020-02-18 The Feinstein Institutes For Medical Research Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US6376190B1 (en) 2000-09-22 2002-04-23 Somalogic, Inc. Modified SELEX processes without purified protein
US6730482B2 (en) 2000-09-22 2004-05-04 Somalogic, Inc. Modified SELEX processes without purified protein
EP2070939A1 (en) 2001-05-25 2009-06-17 Duke University Modulators of pharmacological agents
EP2364990A1 (en) 2001-05-25 2011-09-14 Duke University Modulators of pharmacological agents
US20030143612A1 (en) * 2001-07-18 2003-07-31 Pointilliste, Inc. Collections of binding proteins and tags and uses thereof for nested sorting and high throughput screening
US20080140138A1 (en) * 2002-02-26 2008-06-12 Ivanova Svetlana M Inhibition of inflammatory cytokine production by stimulation of brain muscarinic receptors
US7767803B2 (en) 2002-06-18 2010-08-03 Archemix Corp. Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
US20040249130A1 (en) * 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US20040022727A1 (en) * 2002-06-18 2004-02-05 Martin Stanton Aptamer-toxin molecules and methods for using same
US7960102B2 (en) 2002-07-25 2011-06-14 Archemix Corp. Regulated aptamer therapeutics
WO2004011680A1 (en) 2002-07-25 2004-02-05 Archemix Corp. Regulated aptamer therapeutics
US20060084109A1 (en) * 2002-07-25 2006-04-20 David Epstein Regulated aptamer therapeutics
US20040110235A1 (en) * 2002-07-25 2004-06-10 David Epstein Regulated aptamer therapeutics
US9303262B2 (en) 2002-09-17 2016-04-05 Archemix Llc Methods for identifying aptamer regulators
US20040137010A1 (en) * 2002-09-17 2004-07-15 Charles Wilson Prophylactic and therapeutic HIV aptamers
US8853376B2 (en) 2002-11-21 2014-10-07 Archemix Llc Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US8039443B2 (en) 2002-11-21 2011-10-18 Archemix Corporation Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20040180360A1 (en) * 2002-11-21 2004-09-16 Charles Wilson Multivalent aptamer therapeutics with improved pharmacodynamic properties and methods of making and using the same
US20040253679A1 (en) * 2002-11-21 2004-12-16 David Epstein Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20070009476A1 (en) * 2002-11-21 2007-01-11 Charles Wilson Multivalent aptamer therapeutics with improved pharmacodynamic properties and methods of making and using the same
US10100316B2 (en) 2002-11-21 2018-10-16 Archemix Llc Aptamers comprising CPG motifs
US20050124565A1 (en) * 2002-11-21 2005-06-09 Diener John L. Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20050159351A1 (en) * 2002-11-21 2005-07-21 Dilara Grate Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20040197804A1 (en) * 2002-12-03 2004-10-07 Keefe Anthony D. Method for in vitro selection of 2'-substituted nucleic acids
US20050037394A1 (en) * 2002-12-03 2005-02-17 Keefe Anthony D. Method for in vitro selection of 2'-substituted nucleic acids
US20040253243A1 (en) * 2003-01-21 2004-12-16 David Epstein Aptamer therapeutics useful in ocular pharmacotherapy
US20060216692A1 (en) * 2003-02-04 2006-09-28 Borer Philip N Switchable nucleic acids for diagnostics, screening and molecular electronics
US7718784B2 (en) 2003-02-04 2010-05-18 Syracuse University Switchable nucleic acids for diagnostics, screening and molecular electronics
EP2924119A1 (en) 2003-04-21 2015-09-30 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
EP2623601A2 (en) 2003-04-21 2013-08-07 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US20050250106A1 (en) * 2003-04-24 2005-11-10 David Epstein Gene knock-down by intracellular expression of aptamers
US20040247680A1 (en) * 2003-06-06 2004-12-09 Farokhzad Omid C. Targeted delivery of controlled release polymer systems
US7727969B2 (en) 2003-06-06 2010-06-01 Massachusetts Institute Of Technology Controlled release nanoparticle having bound oligonucleotide for targeted delivery
US20110200593A1 (en) * 2003-08-27 2011-08-18 Ophthotech Corporation Combination Therapy for the Treatment of Ocular Neovascular Disorders
US20100119522A1 (en) * 2003-08-27 2010-05-13 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20050096257A1 (en) * 2003-08-27 2005-05-05 David Shima Combination therapy for the treatment of ocular neovascular disorders
US8685397B2 (en) 2003-08-27 2014-04-01 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US8187597B2 (en) 2003-08-27 2012-05-29 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US8206707B2 (en) 2003-08-27 2012-06-26 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US7759472B2 (en) 2003-08-27 2010-07-20 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20100129364A1 (en) * 2003-08-27 2010-05-27 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20070048248A1 (en) * 2004-02-12 2007-03-01 Claude Benedict Aptamer therapeutics useful in the treatment of complement-related disorders
US7803931B2 (en) 2004-02-12 2010-09-28 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
US7579456B2 (en) 2004-02-12 2009-08-25 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
EP2860251A1 (en) 2004-02-12 2015-04-15 Archemix LLC Aptamer therapeutics useful in the treatment of complement-related disorders
EP3385384A1 (en) 2004-02-12 2018-10-10 Archemix LLC Aptamer therapeutics useful in the treatment of complement-related disorders
EP3736335A1 (en) 2004-02-12 2020-11-11 Archemix LLC Aptamer therapeutics useful in the treatment of complement-related disorders
EP2578683A1 (en) 2004-02-12 2013-04-10 Archemix LLC Aptamer therapeutics useful in the treatment of complement-related disorders
US20060018871A1 (en) * 2004-02-12 2006-01-26 Claude Benedict Aptamer therapeutics useful in the treatment of complement-related disorders
US7538211B2 (en) 2004-02-12 2009-05-26 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
US20060193821A1 (en) * 2004-03-05 2006-08-31 Diener John L Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US20070066550A1 (en) * 2004-03-05 2007-03-22 Diener John L Aptamers to the human IL-12 cytokine family and their use as autoimmune disease therapeutics
US20080249439A1 (en) * 2004-03-25 2008-10-09 The Feinstein Institute For Medical Research Treatment of inflammation by non-invasive stimulation
US20050282906A1 (en) * 2004-03-25 2005-12-22 North Shore-Long Island Jewish Research Institute Neural tourniquet
US8729129B2 (en) 2004-03-25 2014-05-20 The Feinstein Institute For Medical Research Neural tourniquet
US10912712B2 (en) 2004-03-25 2021-02-09 The Feinstein Institutes For Medical Research Treatment of bleeding by non-invasive stimulation
US20080214489A1 (en) * 2004-04-19 2008-09-04 Anthony Dominic Keefe Aptamer-mediated intracellular delivery of oligonucleotides
US7579450B2 (en) 2004-04-26 2009-08-25 Archemix Corp. Nucleic acid ligands specific to immunoglobulin E and their use as atopic disease therapeutics
US20070009907A1 (en) * 2004-04-26 2007-01-11 Paula Burmeister Nucleic acid ligands specific to immunoglobulin E and their use as atopic disease therapeutics
US7829287B2 (en) 2004-08-03 2010-11-09 Syracuse University Method of generating branched and multi-chain nucleic acid switches for ligand detection
US20110059555A1 (en) * 2004-08-03 2011-03-10 Syracuse University Branched and multi-chain nucleic acid switches for sensing and screening
US7521546B2 (en) 2004-08-03 2009-04-21 Syracuse University Branched and multi-chain nucleic acid switches for sensing and screening
US8080379B2 (en) 2004-08-03 2011-12-20 Syracuse University Branched and multi-chain nucleic acid switches for sensing and screening
US20090203025A1 (en) * 2004-08-03 2009-08-13 Syracuse University Branched and multi-chain nucleic acid switches for sensing and screening
US20060029933A1 (en) * 2004-08-03 2006-02-09 Borer Philip N Branched and multi-chain nucleic acid switches for sensing and screening
US20060264369A1 (en) * 2004-09-07 2006-11-23 Diener John L Aptamers to von Willebrand Factor and their use as thrombotic disease therapeutics
US20060183702A1 (en) * 2004-09-07 2006-08-17 Diener John L Aptamers to von Willebrand factor and their use as thrombotic disease therapeutics
US7566701B2 (en) 2004-09-07 2009-07-28 Archemix Corp. Aptamers to von Willebrand Factor and their use as thrombotic disease therapeutics
US7589073B2 (en) 2004-09-07 2009-09-15 Archemix Corp. Aptamers to von Willebrand Factor and their use as thrombotic disease therapeutics
US7998940B2 (en) 2004-09-07 2011-08-16 Archemix Corp. Aptamers to von Willebrand factor and their use as thrombotic disease therapeutics
EP3034089A1 (en) 2004-11-02 2016-06-22 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
EP2436391A2 (en) 2004-11-02 2012-04-04 Archemix LLC Stabilized aptamers to platelet derived growth factor and their use as oncology therapeutics
US11207518B2 (en) 2004-12-27 2021-12-28 The Feinstein Institutes For Medical Research Treating inflammatory disorders by stimulation of the cholinergic anti-inflammatory pathway
US11344724B2 (en) 2004-12-27 2022-05-31 The Feinstein Institutes For Medical Research Treating inflammatory disorders by electrical vagus nerve stimulation
US11913000B2 (en) 2005-02-14 2024-02-27 Iveric Bio, Inc. Aptamer therapeutics useful in the treatment of complement-related disorders
US9617546B2 (en) 2005-02-14 2017-04-11 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
US8236773B2 (en) 2005-02-14 2012-08-07 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
US8436164B2 (en) 2005-02-14 2013-05-07 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
US10947544B2 (en) 2005-02-14 2021-03-16 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
US8946184B2 (en) 2005-02-14 2015-02-03 Archemix Llc Aptamer therapeutics useful in the treatment of complement-related disorders
US20090105172A1 (en) * 2005-03-07 2009-04-23 Diener John L Stabilized Aptamers to PSMA and Their Use as Prostate Cancer Therapeutics
WO2006102540A2 (en) * 2005-03-24 2006-09-28 Syracuse University Elucidation of high affinity, high specificity oligonucleotides and derivatized oligonucleotide sequences for target recognition
WO2006102540A3 (en) * 2005-03-24 2007-01-04 Univ Syracuse Elucidation of high affinity, high specificity oligonucleotides and derivatized oligonucleotide sequences for target recognition
US8921278B2 (en) 2005-03-24 2014-12-30 Syracuse University Method for the discovery of high-affinity, high specificity oligonucleotide and derivatized oligonucleotide sequences for target recognition
US20060257900A1 (en) * 2005-03-24 2006-11-16 Hudson Bruce S Method for the discovery of high-affinity, high specificity oligonucleotide and derivatized oligonucleotide sequences for target recognition
US20090075342A1 (en) * 2005-04-26 2009-03-19 Sharon Cload Metabolic profile directed aptamer medicinal chemistry
US8105813B2 (en) 2005-06-30 2012-01-31 Archemix Corp. Materials and methods for the generation of fully 2′-modified nucleic acid transcripts
US8101385B2 (en) 2005-06-30 2012-01-24 Archemix Corp. Materials and methods for the generation of transcripts comprising modified nucleotides
EP4159220A1 (en) 2006-03-08 2023-04-05 Archemix LLC Complement binding aptamers and anti-c5 agents useful in the treatment of ocular disorders
EP3360582A1 (en) 2006-03-08 2018-08-15 Archemix LLC Complement binding aptamers and anti-c5 agents useful in the treatment of ocular disorders
US20090269356A1 (en) * 2006-03-08 2009-10-29 David Epstein Complement Binding Aptamers and Anti-C5 Agents Useful in the Treatment of Ocular Disorders
EP2596807A1 (en) 2006-03-08 2013-05-29 Archemix LLC Complement binding aptamers and anti-C5 agents useful in the treatment of ocular disorders
EP3034094A1 (en) 2006-03-08 2016-06-22 Archemix LLC Complement binding aptamers and anti-c5 agents useful in the treatment of ocular disorders
US8975388B2 (en) 2007-01-16 2015-03-10 Somalogic, Inc. Method for generating aptamers with improved off-rates
US11111495B2 (en) 2007-01-16 2021-09-07 Somalogic, Inc. Method for generating aptamers with improved off-rates
US10316321B2 (en) 2007-01-16 2019-06-11 Somalogic Inc. Method for generating aptamers with improved off-rates
US7947447B2 (en) 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
EP2629094A1 (en) 2007-01-24 2013-08-21 Carnegie Mellon University Optical biosensors
US20100291562A1 (en) * 2007-04-04 2010-11-18 Michael Adler Method for the detection of an analyte in biological matrix
US20090203766A1 (en) * 2007-06-01 2009-08-13 Archemix Corp. vWF aptamer formulations and methods for use
EP4056711A1 (en) 2007-07-17 2022-09-14 SomaLogic Operating Co., Inc. Method for generating aptamers with improved off-rates
EP2336314A1 (en) 2007-07-17 2011-06-22 Somalogic, Inc. Improved selex and photoselex
EP2933340A1 (en) 2007-07-17 2015-10-21 Somalogic, Inc. Chemically modified aptamers with improved off-rates
EP3284832A2 (en) 2007-07-17 2018-02-21 Somalogic, Inc. Method for generating aptamers with improved off-rates
EP2172566A1 (en) 2007-07-17 2010-04-07 Somalogic, Inc. Method for generating aptameters with improved off-rates
EP2489743A2 (en) 2007-07-17 2012-08-22 Somalogic, Inc. Chemically modified aptamers with improved off-rates
US20090030925A1 (en) * 2007-07-23 2009-01-29 Microsoft Corporation Clustering phylogenetic variation patterns
US8639445B2 (en) 2007-07-23 2014-01-28 Microsoft Corporation Identification of related residues in biomolecular sequences by multiple sequence alignment and phylogenetic analysis
US8391970B2 (en) 2007-08-27 2013-03-05 The Feinstein Institute For Medical Research Devices and methods for inhibiting granulocyte activation by neural stimulation
US20090163437A1 (en) * 2007-10-16 2009-06-25 Regado Biosciences, Inc. Steady-state subcutaneous administration of aptamers
US9910034B2 (en) 2007-11-06 2018-03-06 Ambergen, Inc. Methods and compositions for phototransfer
US10088474B2 (en) 2007-11-06 2018-10-02 Ambergen, Inc. Methods and compositions for phototransfer
US9662490B2 (en) 2008-03-31 2017-05-30 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation and administration of an anti-inflammatory drug
US20090247934A1 (en) * 2008-03-31 2009-10-01 Tracey Kevin J Methods and systems for reducing inflammation by neuromodulation of t-cell activity
US9211409B2 (en) 2008-03-31 2015-12-15 The Feinstein Institute For Medical Research Methods and systems for reducing inflammation by neuromodulation of T-cell activity
US20090275997A1 (en) * 2008-05-01 2009-11-05 Michael Allen Faltys Vagus nerve stimulation electrodes and methods of use
US10329617B2 (en) 2008-05-08 2019-06-25 The Johns Hopkins University Compositions and methods for modulating an immune response
US20100125304A1 (en) * 2008-11-18 2010-05-20 Faltys Michael A Devices and methods for optimizing electrode placement for anti-inflamatory stimulation
US8412338B2 (en) 2008-11-18 2013-04-02 Setpoint Medical Corporation Devices and methods for optimizing electrode placement for anti-inflamatory stimulation
WO2010096388A2 (en) 2009-02-18 2010-08-26 Carnegie Mellon University Quenched dendrimeric dyes for bright detection
US9249306B2 (en) 2009-02-18 2016-02-02 Carnegie Mellon University Quenched dendrimeric dyes for florescence detection
US9211410B2 (en) 2009-05-01 2015-12-15 Setpoint Medical Corporation Extremely low duty-cycle activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
US9849286B2 (en) 2009-05-01 2017-12-26 Setpoint Medical Corporation Extremely low duty-cycle activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
WO2010141771A1 (en) 2009-06-03 2010-12-09 Regado Biosciences, Inc. Nucleic acid modulators of glycoprotein vi
US9174041B2 (en) 2009-06-09 2015-11-03 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US10220203B2 (en) 2009-06-09 2019-03-05 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US10716936B2 (en) 2009-06-09 2020-07-21 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US8886339B2 (en) 2009-06-09 2014-11-11 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
US20100312320A1 (en) * 2009-06-09 2010-12-09 Faltys Michael A Nerve cuff with pocket for leadless stimulator
US9700716B2 (en) 2009-06-09 2017-07-11 Setpoint Medical Corporation Nerve cuff with pocket for leadless stimulator
WO2011006075A1 (en) 2009-07-09 2011-01-13 Somalogic, Inc. Method for generating aptamers with improved off-rates
US8996116B2 (en) 2009-10-30 2015-03-31 Setpoint Medical Corporation Modulation of the cholinergic anti-inflammatory pathway to treat pain or addiction
US9085773B2 (en) 2009-11-03 2015-07-21 Vivonics, Inc. Methods for identifying nucleic acid ligands
US20110104667A1 (en) * 2009-11-03 2011-05-05 Infoscitex Corporation Methods for identifying nucleic acid ligands
US8236570B2 (en) 2009-11-03 2012-08-07 Infoscitex Methods for identifying nucleic acid ligands
US20110104668A1 (en) * 2009-11-03 2011-05-05 Infoscitex Corporation Nucleic acid ligands against infectious prions
US8841429B2 (en) 2009-11-03 2014-09-23 Vivonics, Inc. Nucleic acid ligands against infectious prions
US11051744B2 (en) 2009-11-17 2021-07-06 Setpoint Medical Corporation Closed-loop vagus nerve stimulation
US10384068B2 (en) 2009-12-23 2019-08-20 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US8855767B2 (en) 2009-12-23 2014-10-07 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US8612002B2 (en) 2009-12-23 2013-12-17 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US11110287B2 (en) 2009-12-23 2021-09-07 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US9993651B2 (en) 2009-12-23 2018-06-12 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US20110190849A1 (en) * 2009-12-23 2011-08-04 Faltys Michael A Neural stimulation devices and systems for treatment of chronic inflammation
US9162064B2 (en) 2009-12-23 2015-10-20 Setpoint Medical Corporation Neural stimulation devices and systems for treatment of chronic inflammation
US20110165037A1 (en) * 2010-01-07 2011-07-07 Ismagilov Rustem F Interfaces that eliminate non-specific adsorption, and introduce specific interactions
US9365855B2 (en) 2010-03-03 2016-06-14 Somalogic, Inc. Aptamers to 4-1BB and their use in treating diseases and disorders
US8598140B2 (en) 2010-04-12 2013-12-03 Somalogic, Inc. Aptamers to β-NGF and their use in treating β-NGF mediated diseases and disorders
WO2011150079A1 (en) 2010-05-25 2011-12-01 Carnegie Mellon University Targeted probes of cellular physiology
US9995679B2 (en) 2010-05-25 2018-06-12 Carnegie Mellon University Targeted probes of cellular physiology
US8788034B2 (en) 2011-05-09 2014-07-22 Setpoint Medical Corporation Single-pulse activation of the cholinergic anti-inflammatory pathway to treat chronic inflammation
WO2013034687A1 (en) 2011-09-09 2013-03-14 Universität Bern Verwaltungsdirektion Avibacterium paragallinarum rtx toxin
US9833621B2 (en) 2011-09-23 2017-12-05 Setpoint Medical Corporation Modulation of sirtuins by vagus nerve stimulation
EP3335714A1 (en) 2011-12-30 2018-06-20 Quest Diagnostics Investments Incorporated Aptamers and diagnostic methods for detecting the egf receptor
US10945954B2 (en) 2012-02-06 2021-03-16 President And Fellows Of Harvard College ARRDC1-mediated microvesicles (ARMMS) and uses thereof
US9572983B2 (en) 2012-03-26 2017-02-21 Setpoint Medical Corporation Devices and methods for modulation of bone erosion
US10449358B2 (en) 2012-03-26 2019-10-22 Setpoint Medical Corporation Devices and methods for modulation of bone erosion
EP4219516A2 (en) 2012-07-13 2023-08-02 Wave Life Sciences Ltd. Chiral control
US10942184B2 (en) 2012-10-23 2021-03-09 Caris Science, Inc. Aptamers and uses thereof
EP4170031A1 (en) 2012-10-23 2023-04-26 Caris Science, Inc. Aptamers and uses thereof
US9958448B2 (en) 2012-10-23 2018-05-01 Caris Life Sciences Switzerland Holdings Gmbh Aptamers and uses thereof
WO2014068408A2 (en) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamers and uses thereof
US9939443B2 (en) 2012-12-19 2018-04-10 Caris Life Sciences Switzerland Holdings Gmbh Compositions and methods for aptamer screening
WO2014100434A1 (en) 2012-12-19 2014-06-26 Caris Science, Inc. Compositions and methods for aptamer screening
US11273171B2 (en) 2013-07-12 2022-03-15 Iveric Bio, Inc. Methods for treating or preventing ophthalmological conditions
WO2015031694A2 (en) 2013-08-28 2015-03-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
WO2015075622A1 (en) 2013-11-21 2015-05-28 Inis Biotech Llc Aptamers against the myelin basic protein as neuroprotective agents
US11124822B2 (en) 2014-10-17 2021-09-21 Carnegie Mellon University Enhanced biomolecule detection assays based on tyramide signal amplification and gammaPNA probes
US11311725B2 (en) 2014-10-24 2022-04-26 Setpoint Medical Corporation Systems and methods for stimulating and/or monitoring loci in the brain to treat inflammation and to enhance vagus nerve stimulation
US11001817B2 (en) 2014-10-31 2021-05-11 President And Fellows Of Harvard College Delivery of cargo proteins via ARRDC1-mediated microvesicles (ARMMs)
US11827910B2 (en) 2014-10-31 2023-11-28 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
US10946098B2 (en) 2014-11-17 2021-03-16 Carnegie Mellon University Activatable two-component photosensitizers
US10434177B2 (en) 2014-11-17 2019-10-08 Carnegie Mellon University Activatable two-component photosensitizers
US11406833B2 (en) 2015-02-03 2022-08-09 Setpoint Medical Corporation Apparatus and method for reminding, prompting, or alerting a patient with an implanted stimulator
WO2016145128A1 (en) 2015-03-09 2016-09-15 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11091765B2 (en) 2015-06-29 2021-08-17 Caris Science, Inc. Therapeutic oligonucleotides
WO2017004243A1 (en) 2015-06-29 2017-01-05 Caris Science, Inc. Therapeutic oligonucleotides
US10941176B2 (en) 2015-07-28 2021-03-09 Caris Science, Inc. Therapeutic oligonucleotides
WO2017019918A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
US11725023B2 (en) 2015-07-28 2023-08-15 Caris Science, Inc. Therapeutic oligonucleotides
US10596367B2 (en) 2016-01-13 2020-03-24 Setpoint Medical Corporation Systems and methods for establishing a nerve block
US11278718B2 (en) 2016-01-13 2022-03-22 Setpoint Medical Corporation Systems and methods for establishing a nerve block
US10695569B2 (en) 2016-01-20 2020-06-30 Setpoint Medical Corporation Control of vagal stimulation
US11964150B2 (en) 2016-01-20 2024-04-23 Setpoint Medical Corporation Batteryless implantable microstimulators
US11471681B2 (en) 2016-01-20 2022-10-18 Setpoint Medical Corporation Batteryless implantable microstimulators
US11547852B2 (en) 2016-01-20 2023-01-10 Setpoint Medical Corporation Control of vagal stimulation
US10314501B2 (en) 2016-01-20 2019-06-11 Setpoint Medical Corporation Implantable microstimulators and inductive charging systems
US10583304B2 (en) 2016-01-25 2020-03-10 Setpoint Medical Corporation Implantable neurostimulator having power control and thermal regulation and methods of use
US11383091B2 (en) 2016-01-25 2022-07-12 Setpoint Medical Corporation Implantable neurostimulator having power control and thermal regulation and methods of use
US11332748B2 (en) 2016-03-18 2022-05-17 Caris Science, Inc. Oligonucleotide probes and uses thereof
EP4339288A2 (en) 2016-03-18 2024-03-20 Caris Science, Inc. Oligonucleotide probes and uses thereof
EP3828272A1 (en) 2016-03-18 2021-06-02 Caris Science, Inc. Oligonucleotide probes and uses thereof
US10731166B2 (en) 2016-03-18 2020-08-04 Caris Science, Inc. Oligonucleotide probes and uses thereof
EP4166161A1 (en) 2016-04-14 2023-04-19 Fred Hutchinson Cancer Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
WO2017181110A1 (en) 2016-04-14 2017-10-19 Fred Hutchinson Cancer Research Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
WO2017205686A1 (en) 2016-05-25 2017-11-30 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11293017B2 (en) 2016-05-25 2022-04-05 Caris Science, Inc. Oligonucleotide probes and uses thereof
US11730823B2 (en) 2016-10-03 2023-08-22 President And Fellows Of Harvard College Delivery of therapeutic RNAs via ARRDC1-mediated microvesicles
US11890471B2 (en) 2017-08-14 2024-02-06 Setpoint Medical Corporation Vagus nerve stimulation pre-screening test
US11173307B2 (en) 2017-08-14 2021-11-16 Setpoint Medical Corporation Vagus nerve stimulation pre-screening test
US11660443B2 (en) 2018-04-20 2023-05-30 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via electrical trigeminal nerve stimulation
US11857788B2 (en) 2018-09-25 2024-01-02 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via coordinated trigeminal and vagal nerve stimulation
US11260229B2 (en) 2018-09-25 2022-03-01 The Feinstein Institutes For Medical Research Methods and apparatuses for reducing bleeding via coordinated trigeminal and vagal nerve stimulation
WO2020117914A1 (en) 2018-12-04 2020-06-11 Roche Sequencing Solutions, Inc. Spatially oriented quantum barcoding of cellular targets
US11938324B2 (en) 2020-05-21 2024-03-26 The Feinstein Institutes For Medical Research Systems and methods for vagus nerve stimulation
US11969253B2 (en) 2021-06-02 2024-04-30 Setpoint Medical Corporation Closed-loop vagus nerve stimulation

Also Published As

Publication number Publication date
US7368236B2 (en) 2008-05-06
US5817785A (en) 1998-10-06
US6344318B1 (en) 2002-02-05
US20040241731A1 (en) 2004-12-02
US5595877A (en) 1997-01-21
US6716583B2 (en) 2004-04-06
US20020172962A1 (en) 2002-11-21

Similar Documents

Publication Publication Date Title
US5496938A (en) Nucleic acid ligands to HIV-RT and HIV-1 rev
EP0668931B1 (en) Nucleic acid ligands and methods for producing the same
US5637461A (en) Ligands of HIV-1 TAT protein
EP0786469B1 (en) Methods of use of nucleic acid ligands
US5270163A (en) Methods for identifying nucleic acid ligands
US5705337A (en) Systematic evolution of ligands by exponential enrichment: chemi-SELEX
WO2000079004A1 (en) 2'-fluoropyrimidine anti-calf intestinal phosphatase nucleic acid ligands
JP2009254380A (en) Nucleic acid ligand and method for producing the same
IE84828B1 (en) Methods of use of nucleic acid ligands
Jensen Systematic Evolution Of Ligands By Exponential Enrichment: Chemi-selex
WO1995030775A1 (en) High-affinity ligands of insulin receptor antibodies, tachykinin substance p, hiv integrase and hiv-1 reverse transcriptase

Legal Events

Date Code Title Description
AS Assignment

Owner name: NEXAGEN, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST.;ASSIGNORS:GOLD, LARRY;TUERK, CRAIG;REEL/FRAME:006446/0407

Effective date: 19921207

AS Assignment

Owner name: NEXSTAR PHARMCEUTICALS, INC., COLORADO

Free format text: CHANGE OF NAME;ASSIGNOR:NEXAGEN, INC.;REEL/FRAME:007394/0274

Effective date: 19950221

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: GILEAD SCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEXSTAR PHARMACEUTICALS, INC.;REEL/FRAME:011566/0868

Effective date: 20010123

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12